Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
4.
Am J Transplant ; 21(5): 1902-1909, 2021 05.
Article in English | MEDLINE | ID: mdl-33415805

ABSTRACT

Antibody-mediated deposition of complement membrane attack complexes (MACs) on IFN-γ-primed human endothelial cells (ECs) triggers autocrine/paracrine IL-1ß-mediated EC activation and IL-15 transpresentation to alloreactive effector memory T cells (TEM ), changes that enable ECs to increase T cell proliferation and cytokine release. Here, we report the use of single-cell microchip 32-plex proteomics to more deeply assess the functionality of the activated T cells and dependence upon EC-derived signals. Compared to control ECs, MAC-activated human ECs increase both the frequency and degree of polyfunctionality among both CD4+ and CD8+ -proliferated TEM , assessed as secreted proteins. IFN-γ and TNF-α remain the predominant cytokines made by alloreactive TEM , but a few CD4+ TEM also made IL-4 while more CD8+ TEM made perforin and granzyme B. Increased polyfunctionality was attenuated by treatment of the MAC-activated ECs with anti-IL-15 blocking antibody more effectively than IL-1 receptor blockade. The increased polyfunctionality of T cells resulting from interactions with MAC-activated ECs may further link binding of donor-specific antibody to T cell-mediated allograft pathologies.


Subject(s)
Endothelial Cells , T-Lymphocytes , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Complement System Proteins , Cytokines , Humans , Lymphocyte Activation
5.
J Scleroderma Relat Disord ; 6(1): 111-113, 2021 Feb.
Article in English | MEDLINE | ID: mdl-35382252

ABSTRACT

Morphea, also known as localized scleroderma, is characterized by inflammation and fibrosis of the skin. The exact pathogenesis of morphea is unknown, but generally includes genetic predisposition to autoimmunity combined with an environmental insult. Previous cases have been associated with active Borrelia infection; however, Borrelia infection as a direct cause of morphea was not generalizable to most patients. Within endemic areas, Borrelia burgdorferi is the most common cause of facial nerve paralysis, another autoimmune phenomenon. We report a case of facial morphea in a young man with family history of autoimmune disease who developed morphea in the same location as two previous episodes of Borrelia-induced facial nerve palsy. This case is remarkable because it suggests Borrelia burgdorferi induced loss of local immune tolerance to host antigens, first with facial nerve palsy and followed years later by development of morphea.

6.
Am J Pathol ; 190(6): 1138-1150, 2020 06.
Article in English | MEDLINE | ID: mdl-32194049

ABSTRACT

The complement membrane attack complex (MAC) is classically known as a cytolytic effector of innate and adaptive immunity that forms pores in the plasma membrane of pathogens or targeted cells, leading to osmolysis. Nucleated cells resist MAC-mediated cytolysis by expression of inhibitors that block MAC assembly or by rapid removal of MAC through endocytosis or shedding. In the absence of lysis, MAC may induce intracellular signaling and cell activation, responses implicated in a variety of autoimmune, inflammatory, and transplant disease settings. New discoveries into the structure and biophysical properties of MAC revealed heterogeneous MAC precursors and conformations that provide insights into MAC function. In addition, new mechanisms of MAC-mediated signaling and its contribution to disease pathogenesis have recently come to light. MAC-activated cells have been found to express proinflammatory proteins-often through NF-κB-dependent transcription, assemble inflammasomes, enabling processing, and facilitate secretion of IL-1ß and IL-18, as well as other signaling pathways. These recent insights into the mechanisms of action of MAC provide an updated framework to therapeutic approaches that can target MAC assembly, signaling, and proinflammatory effects in various complement-mediated diseases.


Subject(s)
Adaptive Immunity/physiology , Complement Activation/physiology , Complement Membrane Attack Complex/metabolism , Immunity, Innate/physiology , Animals , Humans , Interleukins/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology
7.
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
8.
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
9.
Circulation ; 141(6): 464-478, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31744330

ABSTRACT

BACKGROUND: Ischemia reperfusion injury (IRI) predisposes to the formation of donor-specific antibodies, a factor contributing to chronic rejection and late allograft loss. METHODS: We describe a mechanism underlying the correlative association between IRI and donor-specific antibodies by using humanized models and patient specimens. RESULTS: IRI induces immunoglobulin M-dependent complement activation on endothelial cells that assembles an NLRP3 (NOD-like receptor pyrin domain-containing protein 3) inflammasome via a Rab5-ZFYVE21-NIK axis and upregulates ICOS-L (inducible costimulator ligand) and PD-L2 (programmed death ligand 2). Endothelial cell-derived interleukin-18 (IL-18) selectively expands a T-cell population (CD4+CD45RO+PD-1hiICOS+CCR2+CXCR5-) displaying features of recently described T peripheral helper cells. This population highly expressed IL-18R1 and promoted donor-specific antibodies in response to IL-18 in vivo. In patients with delayed graft function, a clinical manifestation of IRI, these cells were Ki-67+IL-18R1+ and could be expanded ex vivo in response to IL-18. CONCLUSIONS: IRI promotes elaboration of IL-18 from endothelial cells to selectively expand alloreactive IL-18R1+ T peripheral helper cells in allograft tissues to promote donor-specific antibody formation.


Subject(s)
Human Umbilical Vein Endothelial Cells/immunology , Immunoglobulin M/immunology , Interleukin-18/immunology , Isoantibodies/immunology , Organ Transplantation , Reperfusion Injury/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Delayed Graft Function/immunology , Delayed Graft Function/pathology , Female , Gene Expression Regulation/immunology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammasomes/immunology , Interleukin-18 Receptor alpha Subunit , Mice , Mice, SCID , Reperfusion Injury/pathology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology
10.
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
11.
Sci Rep ; 6: 24697, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27098837

ABSTRACT

Aldosterone-producing adenomas (APAs) vary in phenotype and genotype. Zona glomerulosa (ZG)-like APAs frequently have mutations of an L-type calcium channel (LTCC) CaV1.3. Using a novel antagonist of CaV1.3, compound 8, we investigated the role of CaV1.3 on steroidogenesis in the human adrenocortical cell line, H295R, and in primary human adrenal cells. This investigational drug was compared with the common antihypertensive drug nifedipine, which has 4.5-fold selectivity for the vascular LTCC, CaV1.2, over CaV1.3. In H295R cells transfected with wild-type or mutant CaV1.3 channels, the latter produced more aldosterone than wild-type, which was ameliorated by 100 µM of compound 8. In primary adrenal and non-transfected H295R cells, compound 8 decreased aldosterone production similar to high concentration of nifedipine (100 µM). Selective CaV1.3 blockade may offer a novel way of treating primary hyperaldosteronism, which avoids the vascular side effects of CaV1.2-blockade, and provides targeted treatment for ZG-like APAs with mutations of CaV1.3.


Subject(s)
Aldosterone/metabolism , Calcium Channels, L-Type/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Cell Line , Cells, Cultured , Genotype , Humans , Mutation , Nifedipine/pharmacology , Protein Transport
12.
Protein Eng Des Sel ; 25(12): 827-33, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22961332

ABSTRACT

Building on our previous efforts to generate thermostable chimeric fungal cellobiohydrolase I (CBH I, also known as Cel7A) cellulases by structure-guided recombination, we used FoldX and a 'consensus' sequence approach to identify individual mutations present in the five homologous parent CBH I enzymes which further stabilize the chimeras. Using the FoldX force field, we calculated the effect on ΔG(Folding) of each candidate mutation in a number of CBH I structures and chose those predicted to be stabilizing in multiple structures. With an alignment of 41 CBH I sequences, we also used amino acid frequencies at each candidate position to calculate predicted effects on ΔG(Folding). A combination of mutations chosen using these methods increased the T(50) of the most thermostable chimera by an additional 4.7°C, to yield a CBH I with T(50) of 72.1°C, which is 9.2°C higher than that of the most stable native CBH I, from Talaromyces emersonii. This increased stability resulted in a 10°C increase in the optimal temperature for activity, to 65°C, and a 50% increase in total sugar production from crystalline cellulose at the optimal temperature, compared with native T.emersonii CBH I.


Subject(s)
Cellulose 1,4-beta-Cellobiosidase/chemistry , Cellulose 1,4-beta-Cellobiosidase/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Cellulose 1,4-beta-Cellobiosidase/metabolism , Fungal Proteins/metabolism , Models, Molecular , Mutation , Protein Engineering , Protein Folding , Protein Stability , Talaromyces , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...