Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(37): e2306965120, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37669366

ABSTRACT

Fibrosis is regulated by interactions between immune and mesenchymal cells. However, the capacity of cell types to modulate human fibrosis pathology is poorly understood due to lack of a fully humanized model system. MISTRG6 mice were engineered by homologous mouse/human gene replacement to develop an immune system like humans when engrafted with human hematopoietic stem cells (HSCs). We utilized MISTRG6 mice to model scleroderma by transplantation of healthy or scleroderma skin from a patient with pansclerotic morphea to humanized mice engrafted with unmatched allogeneic HSC. We identified that scleroderma skin grafts contained both skin and bone marrow-derived human CD4 and CD8 T cells along with human endothelial cells and pericytes. Unlike healthy skin, fibroblasts in scleroderma skin were depleted and replaced by mouse fibroblasts. Furthermore, HSC engraftment alleviated multiple signatures of fibrosis, including expression of collagen and interferon genes, and proliferation and activation of human T cells. Fibrosis improvement correlated with reduced markers of T cell activation and expression of human IL-6 by mesenchymal cells. Mechanistic studies supported a model whereby IL-6 trans-signaling driven by CD4 T cell-derived soluble IL-6 receptor complexed with fibroblast-derived IL-6 promoted excess extracellular matrix gene expression. Thus, MISTRG6 mice transplanted with scleroderma skin demonstrated multiple fibrotic responses centered around human IL-6 signaling, which was improved by the presence of healthy bone marrow-derived immune cells. Our results highlight the importance of IL-6 trans-signaling in pathogenesis of scleroderma and the ability of healthy bone marrow-derived immune cells to mitigate disease.


Subject(s)
Basidiomycota , Scleroderma, Localized , Humans , Animals , Mice , Interleukin-6 , Endothelial Cells , Skin , Disease Models, Animal
2.
J Clin Invest ; 130(7): 3437-3452, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32191642

ABSTRACT

Alloantibodies in presensitized transplant candidates deposit complement membrane attack complexes (MACs) on graft endothelial cells (ECs), increasing risk of CD8+ T cell-mediated acute rejection. We recently showed that human ECs endocytose MACs into Rab5+ endosomes, creating a signaling platform that stabilizes NF-κB-inducing kinase (NIK) protein. Endosomal NIK activates both noncanonical NF-κB signaling to synthesize pro-IL-1ß and an NLRP3 inflammasome to process and secrete active IL-1ß. IL-1ß activates ECs, increasing recruitment and activation of alloreactive effector memory CD4+ T (Tem) cells. Here, we report that IFN-γ priming induced nuclear expression of IL-15/IL-15Rα complexes in cultured human ECs and that MAC-induced IL-1ß stimulated translocation of IL-15/IL-15Rα complexes to the EC surface in a canonical NF-κB-dependent process in which IL-15/IL-15Rα transpresentation increased activation and maturation of alloreactive CD8+ Tem cells. Blocking NLRP3 inflammasome assembly, IL-1 receptor, or IL-15 on ECs inhibited the augmented CD8+ Tem cell responses, indicating that this pathway is not redundant. Adoptively transferred alloantibody and mouse complement deposition induced IL-15/IL-15Rα expression by human ECs lining human coronary artery grafts in immunodeficient mice, and enhanced intimal CD8+ T cell infiltration, which was markedly reduced by inflammasome inhibition, linking alloantibody to acute rejection. Inhibiting MAC signaling may similarly limit other complement-mediated pathologies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Complement System Proteins/immunology , Endothelium, Vascular/immunology , Gene Expression Regulation/immunology , Interferon-gamma/immunology , Interleukin-15/immunology , Signal Transduction/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Endothelium, Vascular/cytology , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, SCID , NF-kappa B/immunology , Receptors, Interleukin-15/immunology
3.
Tissue Eng Part A ; 26(5-6): 227-238, 2020 03.
Article in English | MEDLINE | ID: mdl-31672103

ABSTRACT

Multilayered skin substitutes comprising allogeneic cells have been tested for the treatment of nonhealing cutaneous ulcers. However, such nonnative skin grafts fail to permanently engraft because they lack dermal vascular networks important for integration with the host tissue. In this study, we describe the fabrication of an implantable multilayered vascularized bioengineered skin graft using 3D bioprinting. The graft is formed using one bioink containing human foreskin dermal fibroblasts (FBs), human endothelial cells (ECs) derived from cord blood human endothelial colony-forming cells (HECFCs), and human placental pericytes (PCs) suspended in rat tail type I collagen to form a dermis followed by printing with a second bioink containing human foreskin keratinocytes (KCs) to form an epidermis. In vitro, KCs replicate and mature to form a multilayered barrier, while the ECs and PCs self-assemble into interconnected microvascular networks. The PCs in the dermal bioink associate with EC-lined vascular structures and appear to improve KC maturation. When these 3D printed grafts are implanted on the dorsum of immunodeficient mice, the human EC-lined structures inosculate with mouse microvessels arising from the wound bed and become perfused within 4 weeks after implantation. The presence of PCs in the printed dermis enhances the invasion of the graft by host microvessels and the formation of an epidermal rete. Impact Statement Three Dimensional printing can be used to generate multilayered vascularized human skin grafts that can potentially overcome the limitations of graft survival observed in current avascular skin substitutes. Inclusion of human pericytes in the dermal bioink appears to improve both dermal and epidermal maturation.


Subject(s)
Bioprinting/methods , Endothelial Cells/cytology , Fibroblasts/cytology , Keratinocytes/cytology , Pericytes/cytology , Tissue Engineering/methods , Animals , Cells, Cultured , Collagen Type I/metabolism , Endothelial Cells/metabolism , Fibroblasts/metabolism , Flow Cytometry , Humans , Keratinocytes/metabolism , Pericytes/metabolism , Rats , Regenerative Medicine/methods
4.
Circ Res ; 124(12): 1747-1759, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31170059

ABSTRACT

RATIONALE: Complement activation contributes to multiple immune-mediated pathologies. In late allograft failure, donor-specific antibody deposits complement membrane attack complexes (MAC) on graft endothelial cells (ECs), substantially increasing their immunogenicity without causing lysis. Internalized MAC stabilize NIK (NF-κB [nuclear factor kappa-light-chain-enhancer of activated B cells]-inducing kinase) protein on Rab5+MAC+ endosomes, activating noncanonical NF-κB signaling. However, the link to increased immunogenicity is unclear. OBJECTIVE: To identify mechanisms by which alloantibody and internalized MAC activate ECs to enhance their ability to increase T-cell responses. METHODS AND RESULTS: In human EC cultures, internalized MAC also causes NLRP3 (NOD-like receptor family pyrin domain containing 3) translocation from endoplasmic reticulum to Rab5+MAC+NIK+ endosomes followed by endosomal NIK-dependent inflammasome assembly. Cytosolic NIK, stabilized by LIGHT (lymphotoxin-like inducible protein that competes with glycoprotein D for herpesvirus entry on T cells), does not trigger inflammasome assembly, and ATP-triggered inflammasome assembly does not require NIK. IFN-γ (interferon-γ) primes EC responsiveness to MAC by increasing NLRP3, pro-caspase 1, and gasdermin D expression. NIK-activated noncanonical NF-κB signaling induces pro-IL (interleukin)-1ß expression. Inflammasome processed pro-IL-1ß, and gasdermin D results in IL-1ß secretion that increases EC immunogenicity through IL-1 receptor signaling. Activation of human ECs lining human coronary artery grafts in immunodeficient mouse hosts by alloantibody and complement similarly depends on assembly of an NLRP3 inflammasome. Finally, in renal allograft biopsies showing chronic rejection, caspase-1 is activated in C4d+ ECs of interstitial microvessels, supporting the relevance of the cell culture findings. CONCLUSIONS: In response to antibody-mediated complement activation, IFN-γ-primed human ECs internalize MAC, triggering both endosomal-associated NIK-dependent NLRP3 inflammasome assembly and IL-1 synthesis, resulting in autocrine/paracrine IL-1ß-mediated increases in EC immunogenicity. Similar responses may underlie other complement-mediated pathologies.


Subject(s)
Complement Membrane Attack Complex/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Interferon-gamma/pharmacology , Interleukin-1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adult , Cells, Cultured , Endothelium, Vascular/drug effects , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Inflammasomes/metabolism , Male
5.
Nat Commun ; 10(1): 2247, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31113953

ABSTRACT

Complement promotes vascular inflammation in transplant organ rejection and connective tissue diseases. Here we identify ZFYVE21 as a complement-induced Rab5 effector that induces non-canonical NF-κB in endothelial cells (EC). In response to membrane attack complexes (MAC), ZFYVE21 is post-translationally stabilized on MAC+Rab5+ endosomes in a Rab5- and PI(3)P-dependent manner. ZFYVE21 promotes SMURF2-mediated polyubiquitinylation and proteasome-dependent degradation of endosome-associated PTEN to induce vesicular enrichment of PI(3,4,5)P3 and sequential recruitment of activated Akt and NF-κB-inducing kinase (NIK). Pharmacologic alteration of cellular phosphoinositide content with miltefosine reduces ZFYVE21 induction, EC activation, and allograft vasculopathy in a humanized mouse model. ZFYVE21 induction distinctly occurs in response to MAC and is detected in human renal and synovial tissues. Our data identifies ZFYVE21 as a Rab5 effector, defines a Rab5-ZFYVE21-SMURF2-pAkt axis by which it mediates EC activation, and demonstrates a role for this pathway in complement-mediated conditions.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , Graft Rejection/pathology , NF-kappa B/metabolism , Vasculitis/pathology , Allografts/pathology , Animals , Cell Line , Complement Membrane Attack Complex/metabolism , Coronary Vessels/pathology , Coronary Vessels/transplantation , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mice, SCID , Phosphatidylinositol Phosphates/metabolism , Ubiquitin-Protein Ligases/metabolism , rab5 GTP-Binding Proteins/metabolism
6.
Biomaterials ; 183: 128-138, 2018 11.
Article in English | MEDLINE | ID: mdl-30165256

ABSTRACT

Manipulation of human T cell functioning by delivery of macromolecules such as DNA, RNA, or protein is limited, unless the human T cells have been stimulated or electropermeabilized. To achieve successful adaptation and survival of a grafted organ, the alloreactive T cells that induce graft rejection must be regulated. Corticosteroids, calcineurin inhibitors, and mTOR inhibitors, which are systemic immunosuppressants, are currently used for transplantation, with significant side effects. In this study, we demonstrated that a cell-permeable peptide (CPP), dNP2, could efficiently deliver proteins into human CD4 and CD8 T cells. We confirmed regulatory functioning of the cytoplasmic domain of CTLA-4 conjugated with dNP2 (dNP2-ctCTLA-4) in human T cell activation, proliferation, and chemokine receptor expression. We utilized a human skin allograft system in SCID/beige mice to examine whether dNP2-ctCTLA-4 could inhibit allograft rejection by controlling T cell responses. The grafted skin tissue inflammation, allogeneic T cell infiltration, and blood cytokine level was markedly reduced by dNP2-ctCTLA-4, resulting in successful transplantation. In addition, it also inhibited T cell alloresponses against microvessels formed form Bcl-2-transduced human umbilical vein endothelial cells implanted into Balb/c Rag1-/-/IL-2Rγ-/- double knockout (DKO) mice, assessed as reduced T cell infiltration and granzyme B expression. These results collectively suggest that dNP2 peptide conjugation offers a valuable tool for delivering macromolecules like proteins into human T cells, and dNP2-ctCTLA-4 is a novel agent that shows potential in controlling human T cell responses to allow successful adaptation of grafted tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/chemistry , Cell-Penetrating Peptides/chemistry , Graft Rejection/prevention & control , Microvessels/transplantation , Skin Transplantation , T-Lymphocytes/drug effects , Animals , CTLA-4 Antigen/metabolism , Cell Proliferation/drug effects , Cell-Penetrating Peptides/metabolism , Cytokines/blood , Endothelial Cells , Female , Graft Rejection/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , Receptors, Chemokine/metabolism , Skin/immunology , T-Lymphocytes/immunology
7.
JCI Insight ; 3(5)2018 03 08.
Article in English | MEDLINE | ID: mdl-29515027

ABSTRACT

Early acute rejection of human allografts is mediated by circulating alloreactive host effector memory T cells (TEM). TEM infiltration typically occurs across graft postcapillary venules and involves sequential interactions with graft-derived endothelial cells (ECs) and pericytes (PCs). While the role of ECs in allograft rejection has been extensively studied, contributions of PCs to this process are largely unknown. This study aimed to characterize the effects and mechanisms of interactions between human PCs and allogeneic TEM. We report that unstimulated PCs, like ECs, can directly present alloantigen to TEM, but while IFN-γ-activated ECs (γ-ECs) show increased ability to stimulate alloreactive T cells, IFN-γ-activated PCs (γ-PCs) instead suppress TEM proliferation but not cytokine production or signaling. RNA sequencing analysis of PCs, γ-PCs, ECs, and γ-ECs reveal induction of indoleamine 2,3-dioxygenase 1 (IDO1) in γ-PCs to significantly higher levels than in γ-ECs that correlates with tryptophan depletion in vitro. Consistently, shRNA knockdown of IDO1 markedly reduces γ-PC-mediated immunoregulatory effects. Furthermore, human PCs express IDO1 in a skin allograft rejection humanized mouse model and in human renal allografts with acute T cell-mediated rejection. We conclude that immunosuppressive properties of human PCs are not intrinsic but instead result from IFN-γ-induced IDO1-mediated tryptophan depletion.


Subject(s)
Allografts/immunology , Graft Rejection/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/metabolism , Pericytes/immunology , Allografts/blood supply , Allografts/cytology , Animals , Antigen Presentation/immunology , Cell Communication/immunology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Female , Healthy Volunteers , Human Umbilical Vein Endothelial Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-gamma/immunology , Isoantigens/immunology , Mice, SCID , Microvessels/cytology , Microvessels/immunology , Pericytes/metabolism , Primary Cell Culture , RNA, Small Interfering/metabolism , Skin/blood supply , Skin/cytology , Skin/immunology , Skin Transplantation/adverse effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transplantation Chimera , Transplantation, Homologous/adverse effects , Tryptophan/metabolism
8.
Sci Transl Med ; 9(418)2017 11 29.
Article in English | MEDLINE | ID: mdl-29187644

ABSTRACT

Ex vivo normothermic machine perfusion (NMP) is a new clinical strategy to assess and resuscitate organs likely to be declined for transplantation, thereby increasing the number of viable organs available. Short periods of NMP provide a window of opportunity to deliver therapeutics directly to the organ and, in particular, to the vascular endothelial cells (ECs) that constitute the first point of contact with the recipient's immune system. ECs are the primary targets of both ischemia-reperfusion injury and damage from preformed antidonor antibodies, and reduction of perioperative EC injury could have long-term benefits by reducing the intensity of the host's alloimmune response. Using NMP to administer therapeutics directly to the graft avoids many of the limitations associated with systemic drug delivery. We have previously shown that polymeric nanoparticles (NPs) can serve as depots for long-term drug release, but ensuring robust NP accumulation within a target cell type (graft ECs in this case) remains a fundamental challenge of nanomedicine. We show that surface conjugation of an anti-CD31 antibody enhances targeting of NPs to graft ECs of human kidneys undergoing NMP. Using a two-color quantitative microscopy approach, we demonstrate that targeting can enhance EC accumulation by about 5- to 10-fold or higher in discrete regions of the renal vasculature. In addition, our studies reveal that NPs can also nonspecifically accumulate within obstructed regions of the vasculature that are poorly perfused. These quantitative preclinical human studies demonstrate the therapeutic potential for targeted nanomedicines delivered during ex vivo NMP.


Subject(s)
Endothelium/cytology , Endothelium/metabolism , Kidney/cytology , Kidney/metabolism , Endothelial Cells/cytology , Endothelial Cells/physiology , Humans , Nanoparticles , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
9.
J Immunol ; 197(6): 2400-8, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27534549

ABSTRACT

A classical hallmark of acute inflammation is neutrophil infiltration of tissues, a multistep process that involves sequential cell-cell interactions of circulating leukocytes with IL-1- or TNF-activated microvascular endothelial cells (ECs) and pericytes (PCs) that form the wall of the postcapillary venules. The initial infiltrating cells accumulate perivascularly in close proximity to PCs. IL-17, a proinflammatory cytokine that acts on target cells via a heterodimeric receptor formed by IL-17RA and IL-17RC subunits, also promotes neutrophilic inflammation but its effects on vascular cells are less clear. We report that both cultured human ECs and PCs strongly express IL-17RC and, although neither cell type expresses much IL-17RA, PCs express significantly more than ECs. IL-17, alone or synergistically with TNF, significantly alters inflammatory gene expression in cultured human PCs but not ECs. RNA sequencing analysis identifies many IL-17-induced transcripts in PCs encoding proteins known to stimulate neutrophil-mediated immunity. Conditioned media from IL-17-activated PCs, but not ECs, induce pertussis toxin-sensitive neutrophil polarization, likely mediated by PC-secreted chemokines, and they also stimulate neutrophil production of proinflammatory molecules, including TNF, IL-1α, IL-1ß, and IL-8. Furthermore, IL-17-activated PCs, but not ECs, can prolong neutrophil survival by producing G-CSF and GM-CSF, delaying the mitochondrial outer membrane permeabilization and caspase-9 activation. Importantly, neutrophils exhibit enhanced phagocytic capacity after activation by conditioned media from IL-17-treated PCs. We conclude that PCs, not ECs, are the major target of IL-17 within the microvessel wall and that IL-17-activated PCs can modulate neutrophil functions within the perivascular tissue space.


Subject(s)
Endothelium, Vascular/physiology , Interleukin-17/immunology , Neutrophils/immunology , Pericytes/physiology , Receptors, Interleukin-17/immunology , Caspase 9/metabolism , Cells, Cultured , Culture Media , Cytokines/biosynthesis , Cytokines/immunology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Interleukin-17/genetics , Interleukin-17/pharmacology , Neutrophil Infiltration , Neutrophils/physiology , Pericytes/drug effects , Pericytes/immunology , Receptors, Interleukin-17/physiology , Sequence Analysis, RNA , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Venules/cytology , Venules/immunology
10.
Proc Natl Acad Sci U S A ; 112(31): 9686-91, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195760

ABSTRACT

Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.


Subject(s)
Complement Membrane Attack Complex/metabolism , Endosomes/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , rab5 GTP-Binding Proteins/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein , Clathrin/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endocytosis/drug effects , Endosomes/drug effects , Enzyme Stability/drug effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrazones/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , Mice, SCID , Protein Biosynthesis/drug effects , RNA, Small Interfering/metabolism , Secretory Vesicles/drug effects , Secretory Vesicles/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , NF-kappaB-Inducing Kinase
11.
J Exp Med ; 211(3): 395-404, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24516119

ABSTRACT

Recruitment of circulating leukocytes into inflamed tissues depends on adhesion molecules expressed by endothelial cells (ECs). Here we report that rapamycin pretreatment reduced the ability of TNF-treated ECs to capture T cells under conditions of venular flow. This functional change was caused by inhibition of TNF-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and could be mimicked by knockdown of mammalian target of rapamycin (mTOR) or rictor, but not raptor, implicating mTORC2 as the target of rapamycin for this effect. Mechanistically, mTORC2 acts through Akt to repress Raf1-MEK1/2-ERK1/2 signaling, and inhibition of mTORC2 consequently results in hyperactivation of ERK1/2. Increased ERK1/2 activity antagonizes VCAM-1 expression by repressing TNF induction of the transcription factor IRF-1. Preventing activation of ERK1/2 reduced the ability of rapamycin to inhibit TNF-induced VCAM-1 expression. In vivo, rapamycin inhibited mTORC2 activity and potentiated activation of ERK1/2. These changes correlated with reduced endothelial expression of TNF-induced VCAM-1, which was restored via pharmacological inhibition of ERK1/2. Functionally, rapamycin reduced infiltration of leukocytes into renal glomeruli, an effect which was partially reversed by inhibition of ERK1/2. These data demonstrate a novel mechanism by which rapamycin modulates the ability of vascular endothelium to mediate inflammation and identifies endothelial mTORC2 as a potential therapeutic target.


Subject(s)
Endothelial Cells/metabolism , Multiprotein Complexes/antagonists & inhibitors , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/metabolism , Analysis of Variance , Blotting, Western , Cell Adhesion/immunology , Chromatin Immunoprecipitation , DNA Primers/genetics , Flow Cytometry , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 2 , Microscopy, Fluorescence , Multiprotein Complexes/metabolism , Oncogene Protein v-akt/metabolism , Real-Time Polymerase Chain Reaction , T-Lymphocytes/immunology , TOR Serine-Threonine Kinases/metabolism
12.
Circulation ; 128(23): 2504-16, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24045046

ABSTRACT

BACKGROUND: Cardiac allograft vasculopathy is the major cause of late allograft loss after heart transplantation. Cardiac allograft vasculopathy lesions contain alloreactive T cells that secrete interferon-γ, a vasculopathic cytokine, and occur more frequently in patients with donor-specific antibody. Pathological interactions between these immune effectors, representing cellular and humoral immunity, respectively, remain largely unexplored. METHODS AND RESULTS: We used human panel reactive antibody to form membrane attack complexes on allogeneic endothelial cells in vitro and in vivo. Rather than inducing cytolysis, membrane attack complexes upregulated inflammatory genes, enhancing the capacity of endothelial cells to recruit and activate allogeneic interferon-γ--producing CD4(+) T cells in a manner dependent on the activation of noncanonical nuclear factor-κB signaling. Noncanonical nuclear factor-κB signaling was detected in situ within endothelial cells both in renal biopsies from transplantation patients with chronic antibody-mediated rejection and in panel-reactive antibody--treated human coronary artery xenografts in immunodeficient mice. On retransplantation into immunodeficient hosts engrafted with human T cells, panel-reactive antibody--treated grafts recruited more interferon-γ--producing T cells and enhanced cardiac allograft vasculopathy lesion formation. CONCLUSIONS: Alloantibody and complement deposition on graft endothelial cells activates noncanonical nuclear factor-κB signaling, initiating a proinflammatory gene program that enhances alloreactive T cell activation and development of cardiac allograft vasculopathy. Noncanonical nuclear factor-κB signaling in endothelial cells, observed in human allograft specimens and implicated in lesion pathogenesis, may represent a target for new pharmacotherapies to halt the progression of cardiac allograft vasculopathy.


Subject(s)
Complement System Proteins/physiology , Coronary Vessels/immunology , Endothelial Cells/metabolism , Isoantibodies/physiology , NF-kappa B/physiology , Signal Transduction/immunology , T-Lymphocyte Subsets/immunology , Allografts/immunology , Allografts/pathology , Allografts/physiopathology , Animals , Cells, Cultured , Coronary Vessels/pathology , Coronary Vessels/transplantation , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Heterografts/immunology , Human Umbilical Vein Endothelial Cells , Humans , Isoantibodies/blood , Mice , Mice, SCID , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
13.
PLoS One ; 8(2): e55278, 2013.
Article in English | MEDLINE | ID: mdl-23460784

ABSTRACT

OBJECTIVES: Shiga-toxin producing O157:H7 Entero Haemorrhagic E. coli (STEC/EHEC) is one of the most common causes of Haemolytic Uraemic Syndrome (HUS) related to infectious haemorrhagic colitis. Nearly all recommendations on clinical management of EHEC infections refer to this strain. The 2011 outbreak in Northern Europe was the first to be caused by the serotype O104:H4. This EHEC strain was found to carry genetic features of Entero Aggregative E. coli (EAEC) and extended spectrum ß lactamase (ESBL). We report symptoms and complications in patients at one of the most affected centres of the 2011 EHEC O104 outbreak in Northern Germany. METHODS: The courses of patients admitted to our hospital due to bloody diarrhoea with suspected EHEC O104 infection were recorded prospectively. These data include the patients' histories, clinical findings, and complications. RESULTS: EHEC O104 infection was confirmed in 61 patients (female = 37; mean age: 44±2 years). The frequency of HUS was 59% (36/61) in our cohort. An enteric colonisation with co-pathogens was found in 57%. Thirty-one (51%) patients were treated with plasma-separation/plasmapheresis, 16 (26%) with haemodialysis, and 7 (11%) with Eculizumab. Patients receiving antibiotic treatment (n = 37; 61%) experienced no apparent change in their clinical course. Twenty-six (43%) patients suffered from neurological symptoms. One 83-year-old patient died due to comorbidities after HUS was successfully treated. CONCLUSIONS: EHEC O104:H4 infections differ markedly from earlier reports on O157:H7 induced enterocolitis in regard to epidemiology, symptomatology, and frequency of complications. We recommend a standard of practice for clinical monitoring and support the renaming of EHEC O104:H4 syndrome as "EAHEC disease".


Subject(s)
Hemolytic-Uremic Syndrome/microbiology , Hemolytic-Uremic Syndrome/pathology , Hospitalization , Adult , Blood Platelets/pathology , Coinfection/blood , Coinfection/complications , Coinfection/microbiology , Coinfection/virology , Creatinine/blood , Disease Progression , Endoscopy , Enterohemorrhagic Escherichia coli , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Germany/epidemiology , Hemolytic-Uremic Syndrome/diagnostic imaging , Hemolytic-Uremic Syndrome/epidemiology , Hospitalization/statistics & numerical data , Humans , L-Lactate Dehydrogenase/metabolism , Male , Prospective Studies , Time Factors , Ultrasonography
14.
Arterioscler Thromb Vasc Biol ; 32(2): 353-60, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22053072

ABSTRACT

OBJECTIVE: Perioperative nonimmune injuries to an allograft can decrease graft survival. We have developed a model for studying this process using human materials. METHODS AND RESULTS: Human artery segments were transplanted as infrarenal aortic interposition grafts into an immunodeficient mouse host, allowed to "heal in" for 30 days, and then retransplanted into a second mouse host. To induce a reperfusion injury, the healed-in artery segments were incubated for 3 hours under hypoxic conditions ex vivo before retransplantation. To induce immunologic rejection, the animals receiving the retransplanted artery segment were adoptively transferred with human peripheral blood mononuclear cells or purified T cells from a donor allogeneic to the artery 1 week before surgery. To compare rejection of injured versus healthy tissues, these manipulations were combined. Results were analyzed ex vivo by histology, morphometry, immunohistochemistry, and mRNA quantitation or in vivo by ultrasound. Our results showed that reperfusion injury, which otherwise heals with minimal sequelae, intensifies the degree of allogeneic T cell-mediated injury to human artery segments. CONCLUSIONS: We developed a new human-mouse chimeric model demonstrating interactions of reperfusion injury and alloimmunity using human cells and tissues that may be adapted to study other forms of nonimmune injury and other types of adaptive immune responses.


Subject(s)
Adaptive Immunity/physiology , Arteries/immunology , Arteries/transplantation , Chimera/immunology , Reperfusion Injury/physiopathology , T-Lymphocytes/immunology , Adult , Animals , Arteries/pathology , Graft Rejection/immunology , Graft Rejection/physiopathology , Graft Survival/immunology , Graft Survival/physiology , Humans , Mice , Mice, SCID , Models, Animal , T-Lymphocytes/pathology , Transplantation, Homologous
15.
J Biol Chem ; 284(29): 19331-9, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473970

ABSTRACT

Microvascular endothelial cell (EC) expression of tumor necrosis factor receptor (TNFR) 2 is induced in situ by ischemia/reperfusion injury. To assess effects of molecular oxygen on TNFR2 expression, we subjected cultured human dermal microvascular ECs (HDMECs) to hypoxic conditions (1% O(2)) or to hypoxic conditions followed by return to normoxic conditions. TNFR2 mRNA and protein are expressed under normoxic conditions but are rapidly reduced by hypoxia; they fall even further upon reoxygenation but rebound by 6-9 h. TNFR1 expression is unaffected by hypoxia or reoxygenation in these same cells. We identified a potential FOXO3a binding site in the 5' enhancer region of the TNFR2 gene. FOXO3a from normoxic but not hypoxic HDMECs binds an oligonucleotide sequence matching this site, and the endogenous enhancer binds FOXO3a at this site in HDMECs under normoxic but not hypoxic conditions. Unphosphorylated FOXO3a is present in the nucleus of HDMECs under normoxic conditions. Hypoxia leads to FOXO3a phosphorylation at an Akt/protein kinase B target site and subsequent nuclear export; these processes are reversed by reoxygenation and blocked by LY294002, a phosphatidylinositol 3-kinase inhibitor that blocks Akt activation. LY294002 also prevents the hypoxia-mediated decrease in TNFR2 expression. Transiently transfected FOXO3a activates a TNFR2 promoter/reporter construct in HDMECs, whereas small interference RNA knockdown of FOXO3a reduces TNFR2 but not TNFR1 expression under normoxic conditions. Reduction in TNFR2 by small interference RNA sensitizes HDMECs to TNFR1-mediated apoptosis. We conclude that FOXO3a regulates oxygen-dependent changes in expression of TNFR2 in HDMECs, controlling sensitivity to TNF-mediated apoptosis.


Subject(s)
Endothelial Cells/drug effects , Forkhead Transcription Factors/metabolism , Oxygen/pharmacology , Receptors, Tumor Necrosis Factor, Type II/metabolism , Apoptosis/drug effects , Base Sequence , Cell Hypoxia , Cells, Cultured , Chromatin Immunoprecipitation , Chromones/pharmacology , Cycloheximide/pharmacology , Dermis/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression/drug effects , Humans , Immunoblotting , Microscopy, Fluorescence , Molecular Sequence Data , Morpholines/pharmacology , Phosphorylation/drug effects , Protein Synthesis Inhibitors/pharmacology , RNA, Small Interfering/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Necrosis Factor-alpha/pharmacology
16.
Transplantation ; 87(2): 189-97, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19155972

ABSTRACT

BACKGROUND: Nearly half of all infiltrating leukocytes in rejecting human allografts are macrophages, yet, in comparison with T cells, much less is known about the contribution of this cell type to rejection. Our laboratory has previously described models of rejection of human skin or artery grafts in immunodeficient mouse hosts mediated by adoptively transferred allogeneic T cells. However, mature human monocyte/macrophages have consistently failed to engraft in these animals. Here, we describe the introduction of human CD68+ macrophages into irradiated immunodeficient mice by transplantation of enriched CD34+ hematopoietic stem-cells isolated from peripheral blood of G-colony-stimulating factor pretreated adults. METHODS: We investigated strains of immunodeficient mice bearing human tissue grafts (skin and artery) inoculated with 1 x 10(6) human CD34+ adult hematopoietic stem cells, peripheral blood monuclear cells autologous to the CD34 donor, or both for human cell engraftment. RESULTS: In the absence of T cells, CD68+ CD14+ macrophages infiltrate allogeneic human skin but produce little injury or thrombosis. Both responses are enhanced when combined with adoptive transfer of T cells autologous to the hematopoietic stem cells as exemplified by the induction of the macrophage activation marker CD163. CD68+ macrophages also infiltrate allogeneic arterial interposition grafts, producing intimal expansion and calcification in the absence of T cells. CONCLUSIONS: These new models may be used to study the role of human macrophages in transplant rejection and other pathologies in vivo.


Subject(s)
Adult Stem Cells/immunology , Arteries/immunology , Arteries/transplantation , Graft Rejection/immunology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/immunology , Macrophages/immunology , Skin Transplantation/immunology , Skin/immunology , Adoptive Transfer , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Arteries/pathology , Disease Models, Animal , Graft Rejection/pathology , Humans , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/immunology , Lipopolysaccharide Receptors/analysis , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, Cell Surface/analysis , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Transplantation, Homologous , Whole-Body Irradiation
17.
J Exp Med ; 205(13): 3145-58, 2008 Dec 22.
Article in English | MEDLINE | ID: mdl-19075290

ABSTRACT

Interleukin (IL) 1alpha produced by human endothelial cells (ECs), in response to tumor necrosis factor (TNF) or to co-culture with allogeneic T cells in a TNF-dependent manner, can augment the release of cytokines from alloreactive memory T cells in vitro. In a human-mouse chimeric model of artery allograft rejection, ECs lining the transplanted human arteries express IL-1alpha, and blocking IL-1 reduces the extent of human T cell infiltration into the artery intima and selectively inhibits IL-17 production by infiltrating T cells. In human skin grafts implanted on immunodeficient mice, administration of IL-17 is sufficient to induce mild inflammation. In cultured cells, IL-17 acts preferentially on vascular smooth muscle cells rather than ECs to enhance production of proinflammatory mediators, including IL-6, CXCL8, and CCL20. Neutralization of IL-17 does not reduce T cell infiltration into allogeneic human artery grafts, but markedly reduces IL-6, CXCL8, and CCL20 expression and selectively inhibits CCR6(+) T cell accumulation in rejecting arteries. We conclude that graft-derived IL-1 can promote T cell intimal recruitment and IL-17 production during human artery allograft rejection, and suggest that targeting IL-1 in the perioperative transplant period may modulate host alloreactivity.


Subject(s)
Arteries/transplantation , CD4-Positive T-Lymphocytes/immunology , Graft Rejection/immunology , Interleukin-17/immunology , Interleukin-1alpha/immunology , Transplantation, Homologous/immunology , Tunica Intima/immunology , Animals , Arteries/anatomy & histology , Arteries/immunology , CD4-Positive T-Lymphocytes/cytology , Cells, Cultured , Chemokines/immunology , Cytokines/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Humans , Inflammation/immunology , Lymphocyte Activation , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Receptors, CCR6/genetics , Receptors, CCR6/immunology , Skin Transplantation , Tunica Intima/cytology
18.
J Immunol ; 179(7): 4397-404, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17878335

ABSTRACT

The frequency of circulating alloreactive human memory T cells correlates with allograft rejection. Memory T cells may be divided into effector memory (T(EM)) and central memory (T(CM)) cell subsets, but their specific roles in allograft rejection are unknown. We report that CD4+ T(EM) (CD45RO+ CCR7- CD62L-) can be adoptively transferred readily into C.B-17 SCID/bg mice and mediate the destruction of human endothelial cells (EC) in vascularized human skin grafts allogeneic to the T cell donor. In contrast, CD4+ T(CM) (CD45RO+ CCR7+ CD62L+) are inefficiently transferred and do not mediate EC injury. In vitro, CD4+ T(EM) secrete more IFN-gamma within 48 h in response to allogeneic ECs than do T(CM). In contrast, T(EM) and T(CM) secrete comparable amounts of IFN-gamma in response to allogeneic monocytes (Mo). In the same cultures, both T(EM) and T(CM) produce IL-2 and proliferate in response to IFN-gamma-treated allogeneic human EC or Mo, but T(CM) respond more vigorously in both assays. Blockade of LFA-3 strongly inhibits both IL-2 and IFN-gamma secretion by CD4+ T(EM) cultured with allogeneic EC but only minimally inhibits responses to allogeneic Mo. Blockade of CD80 and CD86 strongly inhibits IL-2 but not IFN-gamma production by in response to allogeneic EC or Mo. Transduction of EC to express B7-2 enhances allogeneic T(EM) production of IL-2 but not IFN-gamma. We conclude that human CD4+ T(EM) directly recognize and respond to allogeneic EC in vitro by secreting IFN-gamma and that this response depends on CD2 but not CD28. Consistent with EC activation of effector functions, human CD4+ T(EM) can mediate allogeneic EC injury in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Endothelial Cells/immunology , Animals , Antigen-Presenting Cells/immunology , B7-1 Antigen/immunology , CD4-Positive T-Lymphocytes/metabolism , CD58 Antigens/immunology , Cells, Cultured , Female , Humans , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Mice , Mice, SCID
19.
Proc Natl Acad Sci U S A ; 104(4): 1301-6, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17229850

ABSTRACT

Although antibodies to HLA play a role in the pathogenesis of diseases processes such as rejection of transplanted organs, the precise mechanisms by which antibodies cause tissue injury are not completely understood. We hypothesized that antibodies to host tissues cause inflammation in part by activating endothelial exocytosis of granules that contain prothrombotic mediators such as von Willebrand Factor (VWF) and proinflammatory mediators such as P-selectin. To test this hypothesis, we treated human endothelial cells with murine monoclonal antibody W6/32 to HLA class I and then measured exocytosis by the release of VWF and the externalization of P-selectin. Antibody to HLA activates endothelial exocytosis in a dose-dependent manner over time. The biologically active complement split product, C5a, adds a slight but significant increase to antibody induction of exocytosis. Antibody to HLA alone or with C5a did not damage the cells. Cross-linking of HLA appears to play a role in the ability of antibody to activate exocytosis, because the W6/32 monovalent Fab fragment did not activate VWF release, but the bivalent Fab'2 was effective in triggering exocytosis. To explore the in vivo effects of antibody upon graft injury, we infused W6/32 Fab'2 antibody to human HLA into severe combined immunodeficient/beige mice that had been transplanted with human skin grafts. Antibody to HLA activated exocytosis and inflammation in human skin grafts. Our data show that antibody to host antigens can activate human endothelial cell exocytosis and leukocyte trafficking. By triggering vascular inflammation, antibody activation of exocytosis may play a role in transplant rejection.


Subject(s)
Autoantibodies/immunology , Endothelium, Vascular/cytology , Exocytosis/immunology , HLA Antigens/immunology , Cells, Cultured , Endothelium, Vascular/immunology , Humans , Weibel-Palade Bodies/immunology
20.
J Immunol ; 172(3): 1391-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734714

ABSTRACT

IL-11 can reduce tissue injury in animal models of inflammation but the mechanism(s) is unknown. When C.B-17 SCID/beige mice bearing human skin grafts are injected i.p. with human PBMC allogeneic to the donor skin, infiltrating T cells destroy human microvessels by day 21. Intradermal injection of human IL-11 (500 ng/day) delays the time course of graft microvessel loss without reducing the extent of T cell infiltration. Protective actions of IL-11 are most pronounced on day 15. IL-11 has no effect on T cell activation marker, effector molecule, cytokine expression, or endothelial ICAM-1 expression. IL-11 up-regulates the expression of survivin, a cytoprotective protein, in graft keratinocytes and endothelial cells. Topical application of survivin antisense oligonucleotide down-regulates survivin expression in both cell types and largely abrogates the protective effect of IL-11. We conclude that in this human transplant model, IL-11 exerts a cytoprotective rather than anti-inflammatory or immunomodulatory effect mediated through induction of survivin.


Subject(s)
Cytoprotection/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Interleukin-11/administration & dosage , Microtubule-Associated Proteins/biosynthesis , Skin Transplantation/pathology , Skin/blood supply , Skin/pathology , Adjuvants, Immunologic/administration & dosage , Administration, Topical , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cytoprotection/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Endothelium, Vascular/immunology , Humans , Inhibitor of Apoptosis Proteins , Injections, Intradermal , Interleukin-11/antagonists & inhibitors , Interleukin-11/therapeutic use , Leukocytes, Mononuclear/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Microcirculation/immunology , Microcirculation/metabolism , Microcirculation/pathology , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Neoplasm Proteins , Ointments , Oligonucleotides, Antisense/administration & dosage , Skin/immunology , Skin/metabolism , Skin Transplantation/adverse effects , Skin Transplantation/immunology , Survivin , Transplantation, Homologous/adverse effects , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...