Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Blood Adv ; 8(5): 1105-1115, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38091578

RESUMO

ABSTRACT: Allogeneic hematopoietic cell transplantation (HCT) is a curative therapy for hematological malignancies for which graft-versus-host disease (GVHD) remains a major complication. The use of donor T-regulatory cells (Tregs) to prevent GVHD appears promising, including in our previous evaluation of an engineered graft product (T-reg graft) consisting of the timed, sequential infusion of CD34+ hematopoietic stem cells and high-purity Tregs followed by conventional T cells. However, whether immunosuppressive prophylaxis can be removed from this protocol remains unclear. We report the results of the first stage of an open-label single-center phase 2 study (NCT01660607) investigating T-reg graft in myeloablative HCT of HLA-matched and 9/10-matched recipients. Twenty-four patients were randomized to receive T-reg graft alone (n = 12) or T-reg graft plus single-agent GVHD prophylaxis (n = 12) to determine whether T-reg graft alone was noninferior in preventing acute GVHD. All patients developed full-donor myeloid chimerism. Patients with T-reg graft alone vs with prophylaxis had incidences of grade 3 to 4 acute GVHD of 58% vs 8% (P = .005) and grade 3 to 4 of 17% vs 0% (P = .149), respectively. The incidence of moderate-to-severe chronic GVHD was 28% in the T-reg graft alone arm vs 0% with prophylaxis (P = .056). Among patients with T-reg graft and prophylaxis, CD4+ T-cell-to-Treg ratios were reduced after transplantation, gene expression profiles showed reduced CD4+ proliferation, and the achievement of full-donor T-cell chimerism was delayed. This study indicates that T-reg graft with single-agent tacrolimus is preferred over T-reg graft alone for the prevention of acute GVHD. This trial was registered at www.clinicaltrials.gov as #NCT01660607.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Humanos , Tacrolimo/uso terapêutico , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/prevenção & controle , Doença Enxerto-Hospedeiro/patologia , Imunossupressores/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Doadores de Tecidos
2.
Blood Adv ; 3(18): 2713-2721, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31519647

RESUMO

Peritoneal adhesions are pathological fibroses that ensnare organs after abdominal surgery. This dense connective tissue can cause small bowel obstruction, female infertility, and chronic abdominal pain. The pathogenesis of adhesions is a fibrotic response to tissue damage coordinated between mesothelial cells, fibroblasts, and immune cells. We have previously demonstrated that peritoneal adhesions are a consequence of mechanical injury to the mesothelial layer sustained during surgery. Neutrophils are among the first leukocytes involved in the early response to tissue damage. Here, we show that when subjected to mechanical stress, activated mesothelial cells directly recruit neutrophils and monocytes through upregulation of chemokines such as CXCL1 and monocyte chemoattractant protein 1 (MCP-1). We find that neutrophils within the adhesion sites undergo cell death and form neutrophil extracellular traps (NETosis) that contribute to pathogenesis. Conversely, tissue-resident macrophages were profoundly depleted throughout the disease time course. We show that this is distinct from traditional inflammatory kinetics such as after sham surgery or chemically induced peritonitis, and suggest that adhesions result from a primary difference in inflammatory kinetics. We find that transient depletion of circulating neutrophils significantly decreases adhesion burden, and further recruitment of monocytes with thioglycolate or MCP-1 also improves outcomes. Our findings suggest that the combination of neutrophil depletion and monocyte recruitment is sufficient to prevent adhesion formation, thus providing insight for potential clinical interventions.


Assuntos
Monócitos/metabolismo , Neutrófilos/metabolismo , Aderências Teciduais/metabolismo , Animais , Feminino , Humanos , Camundongos
3.
Cell ; 166(2): 451-467, 2016 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-27419872

RESUMO

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Assuntos
Mesoderma/citologia , Transdução de Sinais , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Coração/crescimento & desenvolvimento , Proteínas de Homeodomínio/metabolismo , Humanos , Mesoderma/metabolismo , Miócitos Cardíacos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Linha Primitiva/citologia , Linha Primitiva/metabolismo , Análise de Célula Única , Somitos/metabolismo , Células-Tronco , Proteínas Supressoras de Tumor/metabolismo , Proteínas Wnt/antagonistas & inibidores , Proteínas Wnt/metabolismo
4.
Proc Natl Acad Sci U S A ; 113(12): E1738-46, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26884166

RESUMO

The specific function of microglia, the tissue resident macrophages of the brain and spinal cord, has been difficult to ascertain because of a lack of tools to distinguish microglia from other immune cells, thereby limiting specific immunostaining, purification, and manipulation. Because of their unique developmental origins and predicted functions, the distinction of microglia from other myeloid cells is critically important for understanding brain development and disease; better tools would greatly facilitate studies of microglia function in the developing, adult, and injured CNS. Here, we identify transmembrane protein 119 (Tmem119), a cell-surface protein of unknown function, as a highly expressed microglia-specific marker in both mouse and human. We developed monoclonal antibodies to its intracellular and extracellular domains that enable the immunostaining of microglia in histological sections in healthy and diseased brains, as well as isolation of pure nonactivated microglia by FACS. Using our antibodies, we provide, to our knowledge, the first RNAseq profiles of highly pure mouse microglia during development and after an immune challenge. We used these to demonstrate that mouse microglia mature by the second postnatal week and to predict novel microglial functions. Together, we anticipate these resources will be valuable for the future study and understanding of microglia in health and disease.


Assuntos
Encéfalo/citologia , Proteínas de Membrana/análise , Microglia/química , Proteínas do Tecido Nervoso/análise , Idoso , Animais , Anticorpos Monoclonais/imunologia , Biomarcadores , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Divisão Celular , Linhagem da Célula , Criança , Endotoxemia/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lipopolissacarídeos/toxicidade , Macrófagos/química , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Microglia/fisiologia , Pessoa de Meia-Idade , Compressão Nervosa , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Traumatismos do Nervo Óptico/patologia , Especificidade de Órgãos , Coelhos , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Análise de Sequência de RNA , Lobo Temporal/metabolismo , Transcriptoma
5.
Stem Cell Reports ; 3(5): 707-15, 2014 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-25418718

RESUMO

Small numbers of hematopoietic stem cells (HSCs) generate large numbers of mature effector cells through the successive amplification of transiently proliferating progenitor cells. HSCs and their downstream progenitors have been extensively characterized based on their cell-surface phenotype and functional activities during transplantation assays. These cells dynamically lose and acquire specific sets of surface markers during differentiation, leading to the identification of markers that allow for more refined separation of HSCs from early hematopoietic progenitors. Here, we describe a marker, CD11A, which allows for the enhanced purification of mouse HSCs. We show through in vivo transplantations that upregulation of CD11A on HSCs denotes the loss of their long-term reconstitution potential. Surprisingly, nearly half of phenotypic HSCs (defined as Lin-KIT(+)SCA-1(+)CD150(+)CD34-) are CD11A(+) and lack long-term self-renewal potential. We propose that CD11A(+)Lin-KIT(+)SCA-1(+)CD150(+)CD34- cells are multipotent progenitors and CD11A-Lin-KIT(+)SCA-1(+)CD150(+)CD34- cells are true HSCs.


Assuntos
Antígeno CD11a/metabolismo , Diferenciação Celular , Proliferação de Células , Células-Tronco Hematopoéticas/metabolismo , Regulação para Cima , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Antígenos Ly/metabolismo , Antígeno CD11a/genética , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores de Superfície Celular/metabolismo , Membro 1 da Família de Moléculas de Sinalização da Ativação Linfocitária
6.
Science ; 341(6141): 88-91, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23722425

RESUMO

CD47 is an antiphagocytic signal that cancer cells employ to inhibit macrophage-mediated destruction. Here, we modified the binding domain of human SIRPα, the receptor for CD47, for use as a CD47 antagonist. We engineered high-affinity SIRPα variants with about a 50,000-fold increased affinity for human CD47 relative to wild-type SIRPα. As high-affinity SIRPα monomers, they potently antagonized CD47 on cancer cells but did not induce macrophage phagocytosis on their own. Instead, they exhibited remarkable synergy with all tumor-specific monoclonal antibodies tested by increasing phagocytosis in vitro and enhancing antitumor responses in vivo. This "one-two punch" directs immune responses against tumor cells while lowering the threshold for macrophage activation, thereby providing a universal method for augmenting the efficacy of therapeutic anticancer antibodies.


Assuntos
Adjuvantes Imunológicos , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Antígenos de Diferenciação/uso terapêutico , Antígeno CD47/imunologia , Neoplasias/terapia , Receptores Imunológicos/uso terapêutico , Animais , Anticorpos Monoclonais Murinos/uso terapêutico , Antígenos de Diferenciação/química , Antígenos de Diferenciação/genética , Linhagem Celular Tumoral , Evolução Molecular Direcionada , Humanos , Imunoterapia , Ativação de Macrófagos , Camundongos , Neoplasias/imunologia , Fagocitose , Receptores Imunológicos/química , Receptores Imunológicos/genética , Rituximab
7.
Proc Natl Acad Sci U S A ; 110(27): 11103-8, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23690610

RESUMO

Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody-mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody-mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8(+))] but decreased priming of OT-II T cells (CD4(+)). The CD4(+) T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3(+)) regulatory T cells. Macrophages following anti-CD47-mediated phagocytosis primed CD8(+) T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antígeno CD47/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/terapia , Macrófagos/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Bloqueadores/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Citotoxicidade Imunológica/genética , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Biol Chem ; 287(51): 43053-62, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23093402

RESUMO

Nitric oxide (NO) signaling regulates key processes in cardiovascular physiology, specifically vasodilation, platelet aggregation, and leukocyte rolling. Soluble guanylate cyclase (sGC), the mammalian NO sensor, transduces an NO signal into the classical second messenger cyclic GMP (cGMP). NO binds to the ferrous (Fe(2+)) oxidation state of the sGC heme cofactor and stimulates formation of cGMP several hundred-fold. Oxidation of the sGC heme to the ferric (Fe(3+)) state desensitizes the enzyme to NO. The heme-oxidized state of sGC has emerged as a potential therapeutic target in the treatment of cardiovascular disease. Here, we investigate the molecular mechanism of NO desensitization and find that sGC undergoes a reductive nitrosylation reaction that is coupled to the S-nitrosation of sGC cysteines. We further characterize the kinetics of NO desensitization and find that heme-assisted nitrosothiol formation of ß1Cys-78 and ß1Cys-122 causes the NO desensitization of ferric sGC. Finally, we provide evidence that the mechanism of reductive nitrosylation is gated by a conformational change of the protein. These results yield insights into the function and dysfunction of sGC in cardiovascular disease.


Assuntos
Guanilato Ciclase/metabolismo , Heme/metabolismo , Ferro/metabolismo , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Alquilação , Animais , Domínio Catalítico , Cisteína/metabolismo , Guanilato Ciclase/química , Humanos , Hidróxidos/metabolismo , Cinética , Proteínas Mutantes/metabolismo , Nitrosação , Nucleotídeos/metabolismo , Oxirredução , Ligação Proteica , Ratos , Receptores Citoplasmáticos e Nucleares/química , Guanilil Ciclase Solúvel , Compostos de Sulfidrila/metabolismo
10.
Biochemistry ; 51(5): 1028-40, 2012 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-22242685

RESUMO

Nitric oxide (NO), the product of the nitric oxide synthase (NOS) reaction, was previously shown to result in S-nitrosation of the NOS Zn(2+)-tetrathiolate and inactivation of the enzyme. To probe the potential physiological significance of NOS S-nitrosation, we determined the inactivation time scale of the inducible NOS isoform (iNOS) and found it directly correlates with an increase in the level of iNOS S-nitrosation. A kinetic model of NOS inactivation in which arginine is treated as a suicide substrate was developed. In this model, NO synthesized at the heme cofactor is partitioned between release into solution (NO release pathway) and NOS S-nitrosation followed by NOS inactivation (inactivation pathway). Experimentally determined progress curves of NO formation were fit to the model. The NO release pathway was perturbed through addition of the NO traps oxymyoglobin (MbO(2)) and ß2 H-NOX, which yielded partition ratios between NO release and inactivation of ~100 at 4 µM MbO(2) and ~22000 at saturating trap concentrations. The results suggest that a portion of the NO synthesized at the heme cofactor reacts with the Zn(2+)-tetrathiolate without being released into solution. Perturbation of the inactivation pathway through addition of the reducing agent GSH or TCEP resulted in a concentration-dependent decrease in the level of iNOS S-nitrosation that directly correlated with protection from iNOS inactivation. iNOS inactivation was most responsive to physiological concentrations of GSH with an apparent K(m) value of 13 mM. NOS turnover that leads to NOS S-nitrosation might be a mechanism for controlling NOS activity, and NOS S-nitrosation could play a role in the physiological generation of nitrosothiols.


Assuntos
Óxido Nítrico Sintase Tipo II/química , Óxido Nítrico Sintase Tipo II/farmacocinética , Animais , Arginina/química , Biotina/química , Biotina/metabolismo , Catálise , Regulação para Baixo/fisiologia , Camundongos , Modelos Moleculares , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Nitrosação , S-Nitrosoglutationa/química , Especificidade por Substrato
11.
Proc Natl Acad Sci U S A ; 106(51): 21602-7, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-20007374

RESUMO

Nitric oxide (NO) regulates a number of essential physiological processes by activating soluble guanylate cyclase (sGC) to produce the second messenger cGMP. The mechanism of NO sensing was previously thought to result exclusively from NO binding to the sGC heme; however, recent studies indicate that heme-bound NO only partially activates sGC and additional NO is involved in the mechanism of maximal NO activation. Furthermore, thiol oxidation of sGC cysteines results in the loss of enzyme activity. Herein the role of cysteines in NO-stimulated sGC activity investigated. We find that the thiol modifying reagent methyl methanethiosulfonate specifically inhibits NO activation of sGC by blocking a non-heme site, which defines a role for sGC cysteine(s) in mediating NO binding. The nature of the NO/cysteine interaction was probed by examining the effects of redox active reagents on NO-stimulated activity. These results show that NO binding to, and dissociation from, the critical cysteine(s) does not involve a change in the thiol redox state. Evidence is provided for non-heme NO in the physiological activation of sGC in context of a primary cell culture of human umbilical vein endothelial cells. These findings have relevance to diseases involving the NO/cGMP signaling pathway.


Assuntos
Cisteína/metabolismo , Guanilato Ciclase/metabolismo , Óxido Nítrico/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Células Cultivadas , GMP Cíclico/metabolismo , Ativação Enzimática , Humanos , Indicadores e Reagentes/química , Oxirredução , Ratos , Guanilil Ciclase Solúvel
12.
Biochemistry ; 48(31): 7519-24, 2009 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-19527054

RESUMO

Soluble guanylate cyclase (sGC) serves as a receptor for the signaling agent nitric oxide (NO). sGC synthesis of cGMP is regulated by NO, GTP, ATP, and allosteric activators such as YC-1. The guanylate cyclase activity and adenylate cyclase activity of full-length sGC and the sGC catalytic domain constructs (alpha1(cat)beta1(cat)) are reported here. ATP is a mixed-type inhibitor of cGMP production for both sGC and alpha1(cat)beta1(cat), indicating that the C-terminus of sGC contains an allosteric nucleotide binding site. YC-1 did not activate alpha1(cat)beta1(cat) or compete with ATP inhibition of cGMP synthesis, which suggests that YC-1 and ATP bind to distinct sites. alpha1(cat)beta1(cat) and NO-stimulated sGC also synthesize cAMP, but this activity is inhibited by ATP via noncompetitive substrate inhibition and by GTP via mixed-type inhibition. Additionally, the adenylate cyclase activity of purified sGC was inhibited by PC12 lysate, suggesting that an intracellular small molecule or protein regulates this activity in vivo.


Assuntos
Trifosfato de Adenosina/química , Guanosina Trifosfato/química , Guanilato Ciclase/metabolismo , Subunidades Proteicas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Trifosfato de Adenosina/antagonistas & inibidores , Trifosfato de Adenosina/fisiologia , Adenilil Ciclases/metabolismo , Regulação Alostérica/fisiologia , Sítio Alostérico/fisiologia , Animais , Domínio Catalítico , Guanosina Trifosfato/metabolismo , Guanosina Trifosfato/fisiologia , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/química , Células PC12 , Conformação Proteica , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/química , Ratos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/química , Guanilil Ciclase Solúvel , Especificidade por Substrato
13.
Proc Natl Acad Sci U S A ; 104(28): 11609-14, 2007 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-17606900

RESUMO

S-nitrosation is a posttranslational, oxidative addition of NO to cysteine residues of proteins that has been proposed as a cGMP-independent signaling pathway [Hess DT, Matsumoto A, Kim SO, Marshall HE, Stamler JS (2005) Nat Rev Mol Cell Biol 6:150-166]. A paradox of S-nitrosation is that only a small set of reactive cysteines are modified in vivo despite the promiscuous reactivity NO exhibits with thiols, precluding the reaction of free NO as the primary mechanism of S-nitrosation. Here we show that a specific transnitrosation reaction between procaspase-3 and thioredoxin-1 (Trx) occurs in cultured human T cells and prevents apoptosis. Trx participation in catalyzing transnitrosation reactions in cells may be general because this protein has numerous protein-protein interactions and plays a key role in cellular redox homeostasis [Powis G, Montfort WR (2001) Annu Rev Pharmacol Toxicol 41:261-295], nitrosothiol content in cells [Haendeler J, Hoffmann J, Tischler V, Berk BC, Zeiher AM, Dimmeler S (2002) Nat Cell Biol 4:743-749], and antiapoptotic signaling.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Precursores Enzimáticos/metabolismo , Nitratos/metabolismo , Tiorredoxinas/metabolismo , Proteínas Reguladoras de Apoptose/fisiologia , Humanos , Células Jurkat , Nitrosação , Transdução de Sinais/fisiologia , Especificidade por Substrato , Tiorredoxinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA