Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Adv Sci (Weinh) ; : e2309976, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38973256

RESUMEN

Efficient and site-specific delivery of therapeutics drugs remains a critical challenge in cancer treatment. Traditional drug nanocarriers such as antibody-drug conjugates are not generally accessible due to their high cost and can lead to serious side effects including life-threatening allergic reactions. Here, these problems are overcome via the engineering of supramolecular agents that are manufactured with an innovative double imprinting approach. The developed molecularly imprinted nanoparticles (nanoMIPs) are targeted toward a linear epitope of estrogen receptor alfa (ERα) and loaded with the chemotherapeutic drug doxorubicin. These nanoMIPs are cost-effective and rival the affinity of commercial antibodies for ERα. Upon specific binding of the materials to ERα, which is overexpressed in most breast cancers (BCs), nuclear drug delivery is achieved via receptor-mediated endocytosis. Consequentially, significantly enhanced cytotoxicity is elicited in BC cell lines overexpressing ERα, paving the way for precision treatment of BC. Proof-of-concept for the clinical use of the nanoMIPs is provided by evaluating their drug efficacy in sophisticated three-dimensional (3D) cancer models, which capture the complexity of the tumor microenvironment in vivo without requiring animal models. Thus, these findings highlight the potential of nanoMIPs as a promising class of novel drug compounds for use in cancer treatment.

2.
Front Immunol ; 15: 1360141, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38361938

RESUMEN

Immunosenescence describes dysregulation of the immune system with ageing manifested in both the innate and adaptive immunity, including changes in T-cell checkpoint signaling. Through complex and nuanced process, T-cells lose excitatory signaling pathways and upregulate their inhibitory signaling, leading to ineffective immune responses that contribute to the formation of the ageing phenotype. Here we expand on the expression, function, and clinical potential of targeting the T-cell checkpoint signaling in age and highlight interventions offering the most benefits to older adults' health. Notably, modifications in vaccination such as with mTOR inhibitors show immediate clinical relevance and good tolerability. Other proposed treatments, including therapies with monoclonal antibodies fail to show clinical efficacy or tolerability needed for implementation at present. Although T-cell co-signaling fits a valuable niche for translational scientists to manage immunosenescence, future study would benefit from the inclusion of older adults with multiple long-term conditions and polypharmacy, ensuring better applicability to actual patients seen in clinical settings.


Asunto(s)
Relevancia Clínica , Inmunosenescencia , Humanos , Anciano , Envejecimiento , Inmunosenescencia/fisiología , Inmunidad Adaptativa , Receptores de Antígenos de Linfocitos T , Linfocitos T
3.
Proc Natl Acad Sci U S A ; 120(18): e2216587120, 2023 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-37098069

RESUMEN

Innate lymphoid cells (ILCs) play a key role in tissue-mediated immunity and can be controlled by coreceptor signaling. Here, we define a subset of ILCs that are Tbet+NK1.1- and are present within the tumor microenvironment (TME). We show programmed death-1 receptor (PD-1) expression on ILCs within TME is found in Tbet+NK1.1- ILCs. PD-1 significantly controlled the proliferation and function of Tbet+NK1.1- ILCs in multiple murine and human tumors. We found tumor-derived lactate enhanced PD-1 expression on Tbet+NK1.1- ILCs within the TME, which resulted in dampened the mammalian target of rapamycin (mTOR) signaling along with increased fatty acid uptake. In line with these metabolic changes, PD-1-deficient Tbet+NK1.1- ILCs expressed significantly increased IFNγ and granzyme B and K. Furthermore, PD-1-deficient Tbet+NK1.1- ILCs contributed toward diminished tumor growth in an experimental murine model of melanoma. These data demonstrate that PD-1 can regulate antitumor responses of Tbet+NK1.1- ILCs within the TME.


Asunto(s)
Linfocitos , Neoplasias , Ratones , Animales , Humanos , Inmunidad Innata , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Microambiente Tumoral , Neoplasias/metabolismo , Apoptosis , Mamíferos/metabolismo
4.
Discov Immunol ; 2(1): kyad003, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38567059

RESUMEN

Innate lymphoid cells (ILCs) are tissue-resident immune cells that have been recently implicated in initiating and driving anti-tumor responses. ILCs are classified into three main groups, namely type 1 ILCs (ILC1), type 2 ILCs, and type 3 ILCs. All three groups have been implicated in either eliciting pro or anti-tumor immune responses in different cancer subtypes with the consensus that ILCs cannot be overlooked within the field of anti-tumor immune responses. In this review, we will specifically expand on the knowledge on ILC1, their characterization, function, and plasticity in anti-cancer immune responses. Within this premise, we will discuss caveats of ILC1 characterization, and expand on the expression and function of immune checkpoint receptors within ILC1 subsets, specifically focusing on the role of programmed cell death-1 receptor in controlling specific ILC1 responses. We summarize that ILC1s are a vital component in initiating anti-tumor responses and can be boosted by checkpoint receptors.

5.
Semin Cancer Biol ; 86(Pt 2): 1045-1055, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34048897

RESUMEN

Programmed cell death-1 (PD-1; CD279) is a cell surface receptor that is expressed in both innate and adaptive immune cells. The role of PD-1 in adaptive immune cells, specifically in CD8+ T cells, has been thoroughly investigated but its significance in other immune cells is yet to be well established. This review will address the role of PD-1 based therapies in enhancing non-CD8+ T cell immune responses within cancer. Specifically, the expression and function of PD-1 in non-CD8+ immune cell compartments such as CD4+ T helper cell subsets, myeloid cells and innate lymphoid cells (ILCs) will be discussed. By understanding the immune cell specific function of PD-1 within tissue resident innate and adaptive immune cells, it will be possible to stratify patients for PD-1 based therapies for both immunogeneic and non-immunogeneic neoplastic disorders. With this knowledge from fundamental and translational studies, PD-1 based therapies can be utilized to enhance T cell independent immune responses in cancers.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Humanos , Microambiente Tumoral , Receptor de Muerte Celular Programada 1 , Inmunoterapia , Inmunidad Innata , Linfocitos/metabolismo , Neoplasias/patología
7.
Nat Cancer ; 1(10): 976-989, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33073241

RESUMEN

Oxidative phosphorylation (OXPHOS) defects caused by somatic mitochondrial DNA (mtDNA) mutations increase with age in human colorectal epithelium and are prevalent in colorectal tumours, but whether they actively contribute to tumorigenesis remains unknown. Here we demonstrate that mtDNA mutations causing OXPHOS defects are enriched during the human adenoma/carcinoma sequence, suggesting they may confer a metabolic advantage. To test this we deleted the tumour suppressor Apc in OXPHOS deficient intestinal stem cells in mice. The resulting tumours were larger than in control mice due to accelerated cell proliferation and reduced apoptosis. We show that both normal crypts and tumours undergo metabolic remodelling in response to OXPHOS deficiency by upregulating the de novo serine synthesis pathway (SSP). Moreover, normal human colonic crypts upregulate the SSP in response to OXPHOS deficiency prior to tumorigenesis. Our data show that age-associated OXPHOS deficiency causes metabolic remodelling that can functionally contribute to accelerated intestinal cancer development.


Asunto(s)
Neoplasias Intestinales , Enfermedades Mitocondriales , Animales , Transformación Celular Neoplásica/genética , ADN Mitocondrial/genética , Neoplasias Intestinales/genética , Ratones , Mitocondrias/genética , Mutación
8.
Eur J Immunol ; 50(6): 770-778, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32383480

RESUMEN

The immune system is tightly regulated by a subset of T cells defined as regulatory T cells (Tregs). Tregs maintain immune homeostasis by restraining unwarranted immune cell activation and effector function. Here, we discuss an important but underappreciated role of proteases in controlling Treg function. Proteases regulate a number of vital processes that determine T cell immune responses and some of them such as furin, ADAM (through regulating LAG receptor), MALT, and asparaginyl endopeptidase are implicated in Treg immunobiology. Targeted protease inhibition, using either small molecule inhibitors or gene deficient mice has demonstrated their specificity in modulating Treg function in experimental murine models. These data further highlight the ability of proteases to specifically regulate Tregs but no other T effector lineages. Taken together, it is apparent that incorporating proteases as targets within Treg cell engineering protocols may enable generation of robust Treg cellular therapeutics. These engineered Tregs may possess enhanced regulatory function along with resistance to lineage deviation in inflammatory disease such as colitis and graft versus host disease. Within this review, we summarize research on the role of proteases in regulating Treg function and discuss the translational potential of harnessing Treg function by targeting protease driven regulatory pathways.


Asunto(s)
Proteínas ADAM/inmunología , Caspasas/inmunología , Colitis , Furina/inmunología , Enfermedad Injerto contra Huésped , Inmunoterapia , Linfocitos T Reguladores/inmunología , Animales , Ingeniería Celular , Colitis/inmunología , Colitis/patología , Colitis/terapia , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Humanos , Ratones , Linfocitos T Reguladores/patología
9.
Methods Mol Biol ; 2121: 1-6, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32147781

RESUMEN

The innate immune system forms a first line of defense against infections and is an essential component in immune defense prior to the generation of sustained adaptive immune responses. Until recently this innate response was believed to be predominantly elicited by cells of myeloid origin. The last 10 years have seen the discovery of an extensive family of innate lymphoid cells (ILCs) characterized by the ability to express cytokines, traditionally associated with activated T effector cells, and lack of expression of antigen receptors.ILCs are enriched at barrier surfaces and rapidly respond to alarmins within the tissue microenvironment. ILCs lack recombinant activating gene (RAG)-dependent rearranged receptors, lack myeloid and dendritic cell lineage defining factors, and possess lymphoid morphology. The first characterized innate lymphoid cells were natural killer cells (NK) cells and lymphoid tissue inducer (LTi) cells. NK cells, defined in 1975), mediate important early antigen independent immune responses. By contrast, LTi cells, defined in 1997, are vital for formation of lymph nodes during embryogenesis. In the past decade, research on ILCs has defined them as important regulators of barrier immunity. However, ILC phenotype and functional characterization is complex and requires highly specific protocols for delineating ILC specific function from other well-characterized immune cells. Within this book, each chapter provides an in-depth protocol that will expand on techniques used by laboratories to study ILC development, characterization and function in mice and humans.


Asunto(s)
Citocinas/metabolismo , Inmunidad Innata , Linfocitos/inmunología , Tejido Linfoide/inmunología , Animales , Linaje de la Célula , Humanos , Células Asesinas Naturales/inmunología , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Ratones , Linfocitos T Colaboradores-Inductores/inmunología
10.
Methods Mol Biol ; 2121: 153-164, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32147794

RESUMEN

Innate lymphoid cells (ILCs) are important for both tissue immunity and tissue homeostasis. They are classified into three groups: Group 1 ILCs include NK cells, which are important in eliciting immunity against intracellular pathogens; group 2 ILCs protect against parasitic helminths; and group 3 ILCs protect against extracellular pathogens. The role of ILCs in cancer immunity remains unclear. In this chapter, we discuss methods for isolating and characterizing tumor-infiltrating ILC subsets within the tumor microenvironment in an experimental murine model of B16 melanoma. The chapter also highlights the expression of PD-1 on the various ILC subsets within the tumor microenvironment.


Asunto(s)
Citometría de Flujo/métodos , Inmunidad Innata , Subgrupos Linfocitarios/citología , Melanoma Experimental/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Subgrupos Linfocitarios/inmunología , Linfocitos/citología , Linfocitos/inmunología , Melanoma Experimental/inducido químicamente , Ratones , Receptor de Muerte Celular Programada 1/metabolismo
11.
Int J Mol Sci ; 20(11)2019 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-31212601

RESUMEN

Programmed cell death-1 (PD-1) is a cell surface receptor that dampens adaptive immune responses. PD-1 is activated by the engagement of its ligands PDL-1 or PDL-2. This results in the inhibition of T cell proliferation, differentiation, cytokine secretion, and cytolytic function. Although a great deal is known about PD-1 mediated regulation of CD4+ and CD8+ T cells, its expression and function in innate lymphoid cells (ILCs) are yet to be fully deciphered. This review summarizes the role of PD-1 in (1) modulating ILC development, (2) ILC function, and (3) PD-1 signaling in ILC. Finally, we explore how PD-1 based immunotherapies may be beneficial in boosting ILC responses in cancer, infections, and other immune-related disorders.


Asunto(s)
Linfocitos/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inmunoterapia , Linfocitos/metabolismo , Receptor de Muerte Celular Programada 1/genética
12.
Immunity ; 49(2): 247-263.e7, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30054205

RESUMEN

CD4+ T cell differentiation into multiple T helper (Th) cell lineages is critical for optimal adaptive immune responses. This report identifies an intrinsic mechanism by which programmed death-1 receptor (PD-1) signaling imparted regulatory phenotype to Foxp3+ Th1 cells (denoted as Tbet+iTregPDL1 cells) and inducible regulatory T (iTreg) cells. Tbet+iTregPDL1 cells prevented inflammation in murine models of experimental colitis and experimental graft versus host disease (GvHD). Programmed death ligand-1 (PDL-1) binding to PD-1 imparted regulatory function to Tbet+iTregPDL1 cells and iTreg cells by specifically downregulating endo-lysosomal protease asparaginyl endopeptidase (AEP). AEP regulated Foxp3 stability and blocking AEP imparted regulatory function in Tbet+iTreg cells. Also, Aep-/- iTreg cells significantly inhibited GvHD and maintained Foxp3 expression. PD-1-mediated Foxp3 maintenance in Tbet+ Th1 cells occurred both in tumor infiltrating lymphocytes (TILs) and during chronic viral infection. Collectively, this report has identified an intrinsic function for PD-1 in maintaining Foxp3 through proteolytic pathway.


Asunto(s)
Cisteína Endopeptidasas/metabolismo , Factores de Transcripción Forkhead/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Diferenciación Celular/inmunología , Células Cultivadas , Colitis/inmunología , Colitis/patología , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/citología , Células TH1/citología
13.
J Exp Med ; 214(6): 1663-1678, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28490441

RESUMEN

Group 2 innate lymphoid cells (ILC-2s) regulate immune responses to pathogens and maintain tissue homeostasis in response to cytokines. Positive regulation of ILC-2s through ICOS has been recently elucidated. We demonstrate here that PD-1 is an important negative regulator of KLRG1+ ILC-2 function in both mice and humans. Increase in KLRG1+ ILC-2 cell numbers was attributed to an intrinsic defect in PD-1 signaling, which resulted in enhanced STAT5 activation. During Nippostrongylus brasiliensis infection, a significant expansion of KLRG1+ ILC-2 subsets occurred in Pdcd1-/- mice and, upon adoptive transfer, Pdcd1-/- KLRG1+ ILC-2s significantly reduced worm burden. Furthermore, blocking PD-1 with an antibody increased KLRG1+ ILC-2 cell number and reduced disease burden. Therefore, PD-1 is required for maintaining the number, and hence function, of KLRG1+ ILC-2s.


Asunto(s)
Inmunidad Innata , Lectinas Tipo C/metabolismo , Linfocitos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Inmunológicos/metabolismo , Transactivadores/metabolismo , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Humanos , Recuento de Linfocitos , Subgrupos Linfocitarios/metabolismo , Ratones Endogámicos C57BL , Nippostrongylus/fisiología , Parásitos/fisiología , Receptor de Muerte Celular Programada 1/deficiencia , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/patología
14.
Haematologica ; 102(8): 1446-1456, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28473623

RESUMEN

CD4+ T-helper subsets drive autoimmune chronic graft-versus-host disease, a major complication after allogeneic bone marrow transplantation. However, it remains unclear how specific T-helper subsets contribute to chronic graft-versus-host disease. T-helper type 1 cells are one of the major disease-mediating T-cell subsets and require interferon-γ signaling and Tbet expression for their function. Regulatory T cells on the other hand can inhibit T-helper type 1 cell-mediated responses. Using an established murine model that isolates the autoimmune component of graft-versus-host disease, we hypothesized that T-helper type 1 cells would restrict FoxP3-driven regulatory T cells. Upon transfer into immune-deficient syngeneic hosts, alloreactive Tbx21-/-CD4+ T cells led to marked increases in FoxP3+ cells and reduced clinical evidence of autoimmunity. To evaluate whether peripheral induction contributed to regulatory T-cell predominance, we adoptively transferred Tbx21-/- T cells that consisted of fate mapping for FoxP3: recipients of flow-purified effector cells that were Foxp3- and Tbx21-/- had enhanced T-regulatory-cell predominance during autoimmune graft-versus-host disease. These data directly demonstrated that peripheral T-regulatory-cell induction was inhibited by Tbet. Finally, Tbx21-/- T-regulatory cells cross-regulated autoimmune wild-type T-effector-cell cytokine production in vivo The Tbet pathway therefore directly impairs T-regulatory-cell reconstitution and is consequently a feasible target in efforts to prevent autoimmune graft-versus-host disease.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Ratones , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Subgrupos de Linfocitos T , Linfocitos T Reguladores/inmunología
15.
Mol Ther ; 24(9): 1655-64, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27401140

RESUMEN

Selective depletion (SD) of alloreactive T cells from allogeneic hematopoeitic stem cell transplants to prevent graft-versus-host disease (GVHD) without compromising immune reconstitution and antitumor responses remains a challenge. Here, we demonstrate a novel SD strategy whereby alloreacting T cells are efficiently deleted ex vivo with adenosine. SD was achieved in human leukocyte antigen (HLA) mismatched cocultures by multiple exposures to 2 mmol/l adenosine over 7 days. Adenosine depleted greater than to 90% of alloproliferating T cells in mismatched, haploidentical, and matched sibling pairs while conserving response to third-party antigens. Alloreactive CD4 and CD8 T cells were targeted for depletion while NK and B cells were preserved. Our novel approach also preserved nonalloreactive naive, central, and effector memory T-cell subsets, Tregs, and notably preserved T-cell responses against DNA viruses that contribute to transplant related mortality after allogeneic hematopoeitic stem cell transplants. Additionally, T cells recognizing leukemia-associated antigens were efficiently generated in vitro from the cell product post-SD. This study is the first to demonstrate that adenosine depletion of alloactivated T cells maintains a complete immune cell profile and recall viral responses. Expansion of tumor antigen-specific subsets postdepletion opens the possibility of generating T-cell products capable of graft-versus-tumor responses without causing GVHD.


Asunto(s)
Adenosina/farmacología , Enfermedad Injerto contra Huésped/prevención & control , Inmunidad , Leucemia/inmunología , Depleción Linfocítica , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Virus/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Neoplasias , Supervivencia Celular/efectos de los fármacos , Enfermedad Injerto contra Huésped/etiología , Haplotipos , Humanos , Leucemia/complicaciones , Leucemia/terapia , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Persona de Mediana Edad , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/metabolismo , Donantes de Tejidos , Adulto Joven
17.
Immunity ; 42(5): 826-38, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25992859

RESUMEN

Interleukin-2 (IL-2) regulates lymphocyte function by signaling through heterodimerization of the IL-2Rß and γc receptor subunits. IL-2 is of considerable therapeutic interest, but harnessing its actions in a controllable manner remains a challenge. Previously, we have engineered an IL-2 "superkine" with enhanced affinity for IL-2Rß. Here, we describe next-generation IL-2 variants that function as "receptor signaling clamps." They retained high affinity for IL-2Rß, inhibiting binding of endogenous IL-2, but their interaction with γc was weakened, attenuating IL-2Rß-γc heterodimerization. These IL-2 analogs acted as partial agonists and differentially affected lymphocytes poised at distinct activation thresholds. Moreover, one variant, H9-RETR, antagonized IL-2 and IL-15 better than blocking antibodies against IL-2Rα or IL-2Rß. Furthermore, this mutein prolonged survival in a model of graft-versus-host disease and blocked spontaneous proliferation of smoldering adult T cell leukemia (ATL) T cells. This receptor-clamping approach might be a general mechanism-based strategy for engineering cytokine partial agonists for therapeutic immunomodulation.


Asunto(s)
Interleucina-2/antagonistas & inhibidores , Ingeniería de Proteínas , Receptores de Interleucina-2/metabolismo , Transducción de Señal/inmunología , Animales , Línea Celular , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped , Humanos , Interleucina-2/química , Interleucina-2/genética , Leucemia-Linfoma de Células T del Adulto/inmunología , Leucemia-Linfoma de Células T del Adulto/fisiopatología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Interleucina-2/química , Factor de Transcripción STAT5/metabolismo , Análisis de Supervivencia
18.
Stem Cells ; 33(4): 1200-12, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25532725

RESUMEN

The use of bone marrow-derived mesenchymal stromal cells (BMSC) in the treatment of alloimmune and autoimmune conditions has generated much interest, yet an understanding of the therapeutic mechanism remains elusive. We therefore explored immune modulation by a clinical-grade BMSC product in a model of human-into-mouse xenogeneic graft-versus-host disease (x-GVHD) mediated by human CD4(+) Th1 cells. BMSC reversed established, lethal x-GVHD through marked inhibition of Th1 cell effector function. Gene marking studies indicated BMSC engraftment was limited to the lung; furthermore, there was no increase in regulatory T cells, thereby suggesting a paracrine mechanism of BMSC action. BMSC recipients had increased serum CD73 expressing exosomes that promoted adenosine accumulation ex vivo. Importantly, immune modulation mediated by BMSC was fully abrogated by pharmacologic therapy with an adenosine A2A receptor antagonist. To investigate the potential clinical relevance of these mechanistic findings, patient serum samples collected pre- and post-BMSC treatment were studied for exosome content: CD73 expressing exosomes promoting adenosine accumulation were detected in post-BMSC samples. In conclusion, BMSC effectively modulate experimental GVHD through a paracrine mechanism that promotes adenosine-based immune suppression.


Asunto(s)
Antagonistas del Receptor de Adenosina A2/farmacología , Células Madre Mesenquimatosas/inmunología , Transducción de Señal/inmunología , Células TH1/inmunología , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Técnicas de Cocultivo , Enfermedad Injerto contra Huésped/inmunología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transducción de Señal/efectos de los fármacos , Células TH1/efectos de los fármacos
19.
Science ; 346(6212): 1000-3, 2014 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-25414314

RESUMEN

Nucleoside reverse transcriptase inhibitors (NRTIs) are mainstay therapeutics for HIV that block retrovirus replication. Alu (an endogenous retroelement that also requires reverse transcriptase for its life cycle)-derived RNAs activate P2X7 and the NLRP3 inflammasome to cause cell death of the retinal pigment epithelium in geographic atrophy, a type of age-related macular degeneration. We found that NRTIs inhibit P2X7-mediated NLRP3 inflammasome activation independent of reverse transcriptase inhibition. Multiple approved and clinically relevant NRTIs prevented caspase-1 activation, the effector of the NLRP3 inflammasome, induced by Alu RNA. NRTIs were efficacious in mouse models of geographic atrophy, choroidal neovascularization, graft-versus-host disease, and sterile liver inflammation. Our findings suggest that NRTIs are ripe for drug repurposing in P2X7-driven diseases.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Inflamasomas/efectos de los fármacos , Inhibidores de la Transcriptasa Inversa/farmacología , Elementos Alu , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Neovascularización Coroidal/tratamiento farmacológico , Modelos Animales de Enfermedad , Atrofia Geográfica/tratamiento farmacológico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Hepatitis/tratamiento farmacológico , Hígado/efectos de los fármacos , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Purinérgicos P2X7/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/fisiología , Inhibidores de la Transcriptasa Inversa/uso terapéutico
20.
Eur J Immunol ; 43(9): 2255-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24037677

RESUMEN

Graft-versus-host disease (GVHD) is a major complication associated with allogeneic bone marrow transplantation (BMT). Recent advances in the treatment of lymphoid malignancies with BMT include exploring mechanisms that can inhibit GVHD while maintaining graft-versus-leukemic (GVL) effects. In this issue of the European Journal of Immunology, Yu et al. [Eur. J. Immunol. 2013.43: 2327-2337] demonstrate efficient separation of GVHD and GVL by abrogating c-Rel in T cells. Intrinsic c-Rel deficiency in T cells resulted in complete protection against GVHD in both major and minor histocompatibility mismatched murine models of BMT. Protection against GVHD was associated with a decreased presence of Th1 and Th17 cells with a concomitant increase in Treg-cell numbers. Interestingly, an intrinsic defect of c-Rel also resulted in decreased expression of the Th1-associated chemokine receptor CXCR3. Finally, the absence of c-Rel maintained GVL effects with significant tumor clearance in murine recipients. These data suggest that specific targeting of the T-cell-specific transcription factor c-Rel can inhibit GVHD while maintaining GVL effects.


Asunto(s)
Trasplante de Médula Ósea/inmunología , Enfermedad Injerto contra Huésped/inmunología , Proteínas Proto-Oncogénicas c-rel/metabolismo , Linfocitos T Reguladores/inmunología , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA