Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(37): e2306965120, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37669366

RESUMEN

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.


Asunto(s)
Basidiomycota , Esclerodermia Localizada , Humanos , Animales , Ratones , Interleucina-6 , Células Endoteliales , Piel , Modelos Animales de Enfermedad
2.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35925682

RESUMEN

Checkpoint inhibitors (CPIs) targeting programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have revolutionized cancer treatment but can trigger autoimmune complications, including CPI-induced diabetes mellitus (CPI-DM), which occurs preferentially with PD-1 blockade. We found evidence of pancreatic inflammation in patients with CPI-DM with shrinkage of pancreases, increased pancreatic enzymes, and in a case from a patient who died with CPI-DM, peri-islet lymphocytic infiltration. In the NOD mouse model, anti-PD-L1 but not anti-CTLA-4 induced diabetes rapidly. RNA sequencing revealed that cytolytic IFN-γ+CD8+ T cells infiltrated islets with anti-PD-L1. Changes in ß cells were predominantly driven by IFN-γ and TNF-α and included induction of a potentially novel ß cell population with transcriptional changes suggesting dedifferentiation. IFN-γ increased checkpoint ligand expression and activated apoptosis pathways in human ß cells in vitro. Treatment with anti-IFN-γ and anti-TNF-α prevented CPI-DM in anti-PD-L1-treated NOD mice. CPIs targeting the PD-1/PD-L1 pathway resulted in transcriptional changes in ß cells and immune infiltrates that may lead to the development of diabetes. Inhibition of inflammatory cytokines can prevent CPI-DM, suggesting a strategy for clinical application to prevent this complication.


Asunto(s)
Diabetes Mellitus , Receptor de Muerte Celular Programada 1 , Animales , Humanos , Mediadores de Inflamación , Ratones , Ratones Endogámicos NOD , Inhibidores del Factor de Necrosis Tumoral
3.
J Clin Invest ; 130(7): 3437-3452, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32191642

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Proteínas del Sistema Complemento/inmunología , Endotelio Vascular/inmunología , Regulación de la Expresión Génica/inmunología , Interferón gamma/inmunología , Interleucina-15/inmunología , Transducción de Señal/inmunología , Animales , Linfocitos T CD8-positivos/citología , Endotelio Vascular/citología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones SCID , FN-kappa B/inmunología , Receptores de Interleucina-15/inmunología
4.
Tissue Eng Part A ; 26(5-6): 227-238, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31672103

RESUMEN

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.


Asunto(s)
Bioimpresión/métodos , Células Endoteliales/citología , Fibroblastos/citología , Queratinocitos/citología , Pericitos/citología , Ingeniería de Tejidos/métodos , Animales , Células Cultivadas , Colágeno Tipo I/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/metabolismo , Citometría de Flujo , Humanos , Queratinocitos/metabolismo , Pericitos/metabolismo , Ratas , Medicina Regenerativa/métodos
5.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31527312

RESUMEN

Tissue engineering may address organ shortages currently limiting clinical transplantation. Off-the-shelf engineered vascularized organs will likely use allogeneic endothelial cells (ECs) to construct microvessels required for graft perfusion. Vasculogenic ECs can be differentiated from committed progenitors (human endothelial colony-forming cells or HECFCs) without risk of mutation or teratoma formation associated with reprogrammed stem cells. Like other ECs, these cells can express both class I and class II major histocompatibility complex (MHC) molecules, bind donor-specific antibody (DSA), activate alloreactive T effector memory cells, and initiate rejection in the absence of donor leukocytes. CRISPR/Cas9-mediated dual ablation of ß2-microglobulin and class II transactivator (CIITA) in HECFC-derived ECs eliminates both class I and II MHC expression while retaining EC functions and vasculogenic potential. Importantly, dually ablated ECs no longer bind human DSA or activate allogeneic CD4+ effector memory T cells and are resistant to killing by CD8+ alloreactive cytotoxic T lymphocytes in vitro and in vivo. Despite absent class I MHC molecules, these ECs do not activate or elicit cytotoxic activity from allogeneic natural killer cells. These data suggest that HECFC-derived ECs lacking MHC molecule expression can be utilized for engineering vascularized grafts that evade allorejection.


Asunto(s)
Aloinjertos/inmunología , Células Endoteliales/inmunología , Rechazo de Injerto/prevención & control , Proteínas Nucleares/genética , Ingeniería de Tejidos/métodos , Transactivadores/genética , Microglobulina beta-2/genética , Aloinjertos/irrigación sanguínea , Aloinjertos/provisión & distribución , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Progenitoras Endoteliales , Femenino , Sangre Fetal/citología , Técnicas de Inactivación de Genes , Rechazo de Injerto/sangre , Rechazo de Injerto/inmunología , Voluntarios Sanos , Humanos , Isoanticuerpos/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/genética , Ratones , Microvasos/citología , Microvasos/inmunología , Microvasos/trasplante , Proteínas Nucleares/inmunología , Trasplante de Órganos/efectos adversos , Trasplante de Órganos/métodos , Cultivo Primario de Células , Transactivadores/inmunología , Microglobulina beta-2/inmunología
6.
Circ Res ; 124(12): 1747-1759, 2019 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-31170059

RESUMEN

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.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Interferón gamma/farmacología , Interleucina-1/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Adulto , Células Cultivadas , Endotelio Vascular/efectos de los fármacos , Femenino , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inflamasomas/metabolismo , Masculino
7.
Nat Commun ; 10(1): 2247, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113953

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , Endosomas/metabolismo , Rechazo de Injerto/patología , FN-kappa B/metabolismo , Vasculitis/patología , Aloinjertos/patología , Animales , Línea Celular , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vasos Coronarios/patología , Vasos Coronarios/trasplante , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones SCID , Fosfatos de Fosfatidilinositol/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
8.
Biomaterials ; 183: 128-138, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30165256

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/química , Péptidos de Penetración Celular/química , Rechazo de Injerto/prevención & control , Microvasos/trasplante , Trasplante de Piel , Linfocitos T/efectos de los fármacos , Animales , Antígeno CTLA-4/metabolismo , Proliferación Celular/efectos de los fármacos , Péptidos de Penetración Celular/metabolismo , Citocinas/sangre , Células Endoteliales , Femenino , Rechazo de Injerto/inmunología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/trasplante , Humanos , Activación de Linfocitos , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Receptores de Quimiocina/metabolismo , Piel/inmunología , Linfocitos T/inmunología
9.
JCI Insight ; 3(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29515027

RESUMEN

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.


Asunto(s)
Aloinjertos/inmunología , Rechazo de Injerto/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Pericitos/inmunología , Aloinjertos/irrigación sanguínea , Aloinjertos/citología , Animales , Presentación de Antígeno/inmunología , Comunicación Celular/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Endotelio Vascular/citología , Femenino , Voluntarios Sanos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón gamma/inmunología , Isoantígenos/inmunología , Ratones SCID , Microvasos/citología , Microvasos/inmunología , Pericitos/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Piel/irrigación sanguínea , Piel/citología , Piel/inmunología , Trasplante de Piel/efectos adversos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Quimera por Trasplante , Trasplante Homólogo/efectos adversos , Triptófano/metabolismo
10.
Sci Transl Med ; 9(418)2017 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-29187644

RESUMEN

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.


Asunto(s)
Endotelio/citología , Endotelio/metabolismo , Riñón/citología , Riñón/metabolismo , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Nanopartículas , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
11.
J Immunol ; 197(6): 2400-8, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27534549

RESUMEN

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.


Asunto(s)
Endotelio Vascular/fisiología , Interleucina-17/inmunología , Neutrófilos/inmunología , Pericitos/fisiología , Receptores de Interleucina-17/inmunología , Caspasa 9/metabolismo , Células Cultivadas , Medios de Cultivo , Citocinas/biosíntesis , Citocinas/inmunología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/inmunología , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Factor Estimulante de Colonias de Granulocitos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interleucina-17/genética , Interleucina-17/farmacología , Infiltración Neutrófila , Neutrófilos/fisiología , Pericitos/efectos de los fármacos , Pericitos/inmunología , Receptores de Interleucina-17/fisiología , Análisis de Secuencia de ARN , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Vénulas/citología , Vénulas/inmunología
12.
Proc Natl Acad Sci U S A ; 112(31): 9686-91, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195760

RESUMEN

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.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Endosomas/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab5/metabolismo , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Clatrina/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hidrazonas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones SCID , Biosíntesis de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Transducción de Señal/efectos de los fármacos , Factor 3 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Quinasa de Factor Nuclear kappa B
13.
J Exp Med ; 211(3): 395-404, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24516119

RESUMEN

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.


Asunto(s)
Células Endoteliales/metabolismo , Complejos Multiproteicos/antagonistas & inhibidores , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Molécula 1 de Adhesión Celular Vascular/metabolismo , Análisis de Varianza , Western Blotting , Adhesión Celular/inmunología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Immunoblotting , Diana Mecanicista del Complejo 2 de la Rapamicina , Microscopía Fluorescente , Complejos Multiproteicos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo
14.
Circulation ; 128(23): 2504-16, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24045046

RESUMEN

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.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Vasos Coronarios/inmunología , Células Endoteliales/metabolismo , Isoanticuerpos/fisiología , FN-kappa B/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Aloinjertos/inmunología , Aloinjertos/patología , Aloinjertos/fisiopatología , Animales , Células Cultivadas , Vasos Coronarios/patología , Vasos Coronarios/trasplante , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Xenoinjertos/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isoanticuerpos/sangre , Ratones , Ratones SCID , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
15.
PLoS One ; 8(2): e55278, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23460784

RESUMEN

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".


Asunto(s)
Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Hospitalización , Adulto , Plaquetas/patología , Coinfección/sangre , Coinfección/complicaciones , Coinfección/microbiología , Coinfección/virología , Creatinina/sangre , Progresión de la Enfermedad , Endoscopía , Escherichia coli Enterohemorrágica , Heces/microbiología , Femenino , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Alemania/epidemiología , Síndrome Hemolítico-Urémico/diagnóstico por imagen , Síndrome Hemolítico-Urémico/epidemiología , Hospitalización/estadística & datos numéricos , Humanos , L-Lactato Deshidrogenasa/metabolismo , Masculino , Estudios Prospectivos , Factores de Tiempo , Ultrasonografía
16.
Arterioscler Thromb Vasc Biol ; 32(2): 353-60, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22053072

RESUMEN

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.


Asunto(s)
Inmunidad Adaptativa/fisiología , Arterias/inmunología , Arterias/trasplante , Quimera/inmunología , Daño por Reperfusión/fisiopatología , Linfocitos T/inmunología , Adulto , Animales , Arterias/patología , Rechazo de Injerto/inmunología , Rechazo de Injerto/fisiopatología , Supervivencia de Injerto/inmunología , Supervivencia de Injerto/fisiología , Humanos , Ratones , Ratones SCID , Modelos Animales , Linfocitos T/patología , Trasplante Homólogo
17.
Sci Rep ; 1: 158, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22355673

RESUMEN

The accessibility of skin makes it an ideal target organ for nucleic acid-based therapeutics; however, effective patient-friendly delivery remains a major obstacle to clinical utility. A variety of limited and inefficient methods of delivering nucleic acids to keratinocytes have been demonstrated; further advances will require well-characterized reagents, rapid noninvasive assays of delivery, and well-developed skin model systems. Using intravital fluorescence and bioluminescence imaging and a standard set of reporter plasmids we demonstrate transfection of cells in mouse and human xenograft skin using intradermal injection and two microneedle array delivery systems. Reporter gene expression could be detected in individual keratinocytes, in real-time, in both mouse skin as well as human skin xenografts. These studies revealed that non-invasive intravital imaging can be used as a guide for developing gene delivery tools, establishing a benchmark for comparative testing of nucleic acid skin delivery technologies.


Asunto(s)
Queratinocitos/metabolismo , Plásmidos/administración & dosificación , Animales , Sistemas de Liberación de Medicamentos , Epidermis/metabolismo , Femenino , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes , Ratones , Microinyecciones , Microscopía Fluorescente , Plásmidos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trasplante de Piel , Transfección , Trasplante Heterólogo
18.
Hum Immunol ; 70(10): 790-802, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19524633

RESUMEN

Immunodeficient mice bearing components of a human immune system present a novel approach for studying human immune responses. We investigated the number, phenotype, developmental kinetics, and function of developing human immune cells following transfer of CD34(+) hematopoietic stem cell (HSC) preparations originating from second trimester human fetal liver (HFL), umbilical cord blood (UCB), or granulocyte colony-stimulating factor-mobilized adult blood (G-CSF-AB) delivered via intrahepatic injection into sublethally irradiated neonatal NOD-scid/gammac(-/-), Balb/c-Rag1(-/-)gammac(-/-), and C.B-17-scid/bg mice. HFL and UCB HSC provided the greatest number and breadth of developing cells. NOD-scid/gammac(-/-) and Balb/c-Rag1(-/-)gammac(-/-) harbored human B and dendritic cells as well as human platelets in peripheral blood, whereas NOD-scid/gammac(-/-) mice harbored higher levels of human T cells. NOD-scid/gammac(-/-) mice engrafted with HFL CD34(+) HSC demonstrated human immunological competence evidenced by white pulp expansion and increases in total human immunoglobulin following immunization with T-dependent antigens and delayed-type hypersensitivity-infiltrating leukocytes in response to antigenic challenge. In conclusion, we describe an encouraging base system for studying human hematopoietic lineage development and function utilizing human HFL or UCB HSC-engrafted NOD-scid/gammac(-/-) mice that is well suited for future studies toward the development of a fully competent humanized mouse model.


Asunto(s)
Sangre Fetal/inmunología , Células Madre Hematopoyéticas/inmunología , Hígado/inmunología , Subgrupos Linfocitarios/inmunología , Animales , Factor Estimulante de Colonias de Granulocitos/inmunología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Movilización de Célula Madre Hematopoyética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hemocianinas/inmunología , Humanos , Hipersensibilidad Tardía/inmunología , Hipersensibilidad Tardía/patología , Inmunoglobulinas/sangre , Subtipo H5N1 del Virus de la Influenza A/inmunología , Hígado/embriología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Subgrupos Linfocitarios/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Dosis de Radiación , Bazo/citología , Bazo/inmunología , Timo/citología , Timo/inmunología , Irradiación Corporal Total
19.
J Biol Chem ; 284(29): 19331-9, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19473970

RESUMEN

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.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Oxígeno/farmacología , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Apoptosis/efectos de los fármacos , Secuencia de Bases , Hipoxia de la Célula , Células Cultivadas , Inmunoprecipitación de Cromatina , Cromonas/farmacología , Cicloheximida/farmacología , Dermis/citología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Microscopía Fluorescente , Datos de Secuencia Molecular , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , ARN Interferente Pequeño/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Factor de Necrosis Tumoral alfa/farmacología
20.
Transplantation ; 87(2): 189-97, 2009 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-19155972

RESUMEN

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.


Asunto(s)
Células Madre Adultas/inmunología , Arterias/inmunología , Arterias/trasplante , Rechazo de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/inmunología , Macrófagos/inmunología , Trasplante de Piel/inmunología , Piel/inmunología , Traslado Adoptivo , Animales , Antígenos CD/análisis , Antígenos CD34/análisis , Antígenos de Diferenciación Mielomonocítica/análisis , Arterias/patología , Modelos Animales de Enfermedad , Rechazo de Injerto/patología , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Receptores de Lipopolisacáridos/análisis , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptores de Superficie Celular/análisis , Piel/patología , Linfocitos T/inmunología , Linfocitos T/trasplante , Trasplante Homólogo , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...