Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Brain ; 147(2): 566-589, 2024 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-37776513

RESUMO

Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.


Assuntos
Malária Cerebral , Camundongos , Humanos , Animais , Malária Cerebral/patologia , Malária Cerebral/prevenção & controle , Células Endoteliais/patologia , Encéfalo/patologia , Barreira Hematoencefálica/patologia , Linfócitos T CD8-Positivos , Endotélio/patologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
2.
Blood ; 139(26): 3708-3721, 2022 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-35090171

RESUMO

Pivotal clinical trials of B-cell maturation antigen-targeted chimeric antigen receptor T (CART)-cell therapy in patients with relapsed/refractory multiple myeloma (MM) resulted in remarkable initial responses, which led to a recent US Food and Drug Administration approval. Despite the success of this therapy, durable remissions continue to be low, and the predominant mechanism of resistance is loss of CART cells and inhibition by the tumor microenvironment (TME). MM is characterized by an immunosuppressive TME with an abundance of cancer-associated fibroblasts (CAFs). Using MM models, we studied the impact of CAFs on CART-cell efficacy and developed strategies to overcome CART-cell inhibition. We showed that CAFs inhibit CART-cell antitumor activity and promote MM progression. CAFs express molecules such as fibroblast activation protein and signaling lymphocyte activation molecule family-7, which are attractive immunotherapy targets. To overcome CAF-induced CART-cell inhibition, CART cells were generated targeting both MM cells and CAFs. This dual-targeting CART-cell strategy significantly improved the effector functions of CART cells. We show for the first time that dual targeting of both malignant plasma cells and the CAFs within the TME is a novel strategy to overcome resistance to CART-cell therapy in MM.


Assuntos
Fibroblastos Associados a Câncer , Mieloma Múltiplo , Medula Óssea , Fibroblastos Associados a Câncer/patologia , Terapia Baseada em Transplante de Células e Tecidos , Fibroblastos , Humanos , Imunoterapia Adotiva/métodos , Mieloma Múltiplo/patologia , Microambiente Tumoral
3.
Brain Behav Immun ; 112: 51-76, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37236326

RESUMO

The contribution of circulating verses tissue resident memory T cells (TRMs) to clinical neuropathology is an enduring question due to a lack of mechanistic insights. The prevailing view is TRMs are protective against pathogens in the brain. However, the extent to which antigen-specific TRMs induce neuropathology upon reactivation is understudied. Using the described phenotype of TRMs, we found that brains of naïve mice harbor populations of CD69+ CD103- T cells. Notably, numbers of CD69+ CD103- TRMs rapidly increase following neurological insults of various origins. This TRM expansion precedes infiltration of virus antigen-specific CD8 T cells and is due to proliferation of T cells within the brain. We next evaluated the capacity of antigen-specific TRMs in the brain to induce significant neuroinflammation post virus clearance, including infiltration of inflammatory myeloid cells, activation of T cells in the brain, microglial activation, and significant blood brain barrier disruption. These neuroinflammatory events were induced by TRMs, as depletion of peripheral T cells or blocking T cell trafficking using FTY720 did not change the neuroinflammatory course. Depletion of all CD8 T cells, however, completely abrogated the neuroinflammatory response. Reactivation of antigen-specific TRMs in the brain also induced profound lymphopenia within the blood compartment. We have therefore determined that antigen-specific TRMs can induce significant neuroinflammation, neuropathology, and peripheral immunosuppression. The use of cognate antigen to reactivate CD8 TRMs enables us to isolate the neuropathologic effects induced by this cell type independently of other branches of immunological memory, differentiating this work from studies employing whole pathogen re-challenge. This study also demonstrates the capacity for CD8 TRMs to contribute to pathology associated with neurodegenerative disorders and long-term complications associated with viral infections. Understanding functions of brain TRMs is crucial in investigating their role in neurodegenerative disorders including MS, CNS cancers, and long-term complications associated with viral infections including COVID-19.


Assuntos
COVID-19 , Viroses , Camundongos , Animais , Células T de Memória , Doenças Neuroinflamatórias , Linfócitos T CD8-Positivos , Encéfalo , Memória Imunológica
4.
J Immunol ; 205(5): 1228-1238, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32737149

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) infection of the CNS is cleared in C57BL/6 mice by a CD8 T cell response restricted by the MHC class I molecule H-2Db The identity and function of the APC(s) involved in the priming of this T cell response is (are) poorly defined. To address this gap in knowledge, we developed an H-2Db LoxP-transgenic mouse system using otherwise MHC class I-deficient C57BL/6 mice, thereby conditionally ablating MHC class I-restricted Ag presentation in targeted APC subpopulations. We observed that CD11c+ APCs are critical for early priming of CD8 T cells against the immunodominant TMEV peptide VP2121-130 Loss of H-2Db on CD11c+ APCs mitigates the CD8 T cell response, preventing early viral clearance and immunopathology associated with CD8 T cell activity in the CNS. In contrast, animals with H-2Db-deficient LysM+ APCs retained early priming of Db:VP2121-130 epitope-specific CD8 T cells, although a modest reduction in immune cell entry into the CNS was observed. This work establishes a model enabling the critical dissection of H-2Db-restricted Ag presentation to CD8 T cells, revealing cell-specific and temporal features involved in the generation of CD8 T cell responses. Employing this novel system, we establish CD11c+ cells as pivotal to the establishment of acute antiviral CD8 T cell responses against the TMEV immunodominant epitope VP2121-130, with functional implications both for T cell-mediated viral control and immunopathology.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Cardiovirus/imunologia , Genes MHC Classe I/imunologia , Antígenos H-2/imunologia , Theilovirus/imunologia , Animais , Apresentação de Antígeno , Proteínas do Capsídeo/imunologia , Epitopos de Linfócito T/imunologia , Epitopos Imunodominantes/imunologia , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
5.
Mol Ther ; 29(4): 1529-1540, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33388419

RESUMO

Chimeric antigen receptor (CAR) T cell therapy has yielded unprecedented outcomes in some patients with hematological malignancies; however, inhibition by the tumor microenvironment has prevented the broader success of CART cell therapy. We used chronic lymphocytic leukemia (CLL) as a model to investigate the interactions between the tumor microenvironment and CART cells. CLL is characterized by an immunosuppressive microenvironment, an abundance of systemic extracellular vesicles (EVs), and a relatively lower durable response rate to CART cell therapy. In this study, we characterized plasma EVs from untreated CLL patients and identified their leukemic cell origin. CLL-derived EVs were able to induce a state of CART cell dysfunction characterized by phenotypical, functional, and transcriptional changes of exhaustion. We demonstrate that, specifically, PD-L1+ CLL-derived EVs induce CART cell exhaustion. In conclusion, we identify an important mechanism of CART cell exhaustion induced by EVs from CLL patients.


Assuntos
Antígeno B7-H1/sangue , Leucemia Linfocítica Crônica de Células B/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Vesículas Extracelulares/genética , Vesículas Extracelulares/imunologia , Feminino , Humanos , Imunoterapia Adotiva/métodos , Leucemia Linfocítica Crônica de Células B/sangue , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Masculino , Receptores de Antígenos de Linfócitos T/sangue , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Microambiente Tumoral/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 116(8): 3136-3145, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30728302

RESUMO

Successful efforts to activate T cells capable of recognizing weak cancer-associated self-antigens have employed altered peptide antigens to activate T cell responses capable of cross-reacting on native tumor-associated self. A limitation of this approach is the requirement for detailed knowledge about the altered self-peptide ligands used in these vaccines. In the current study we considered allorecognition as an approach for activating CTL capable of recognizing weak or self-antigens in the context of self-MHC. Nonself antigen-presenting molecules typically contain polymorphisms that influence interactions with the bound peptide and TCR interface. Recognition of these nonself structures results in peptide-dependent alloimmunity. Alloreactive T cells target their inducing alloantigens as well as third-party alloantigens but generally fail to target self-antigens. Certain residues located on the alpha-1/2 domains of class I antigen-presenting molecules primarily interface with TCR. These residues are more conserved within and across species than are residues that determine peptide antigen binding properties. Class I variants designed with amino acid substitutions at key positions within the conserved helical structures are shown to provide strong activating signals to alloreactive CD8 T cells while avoiding changes in naturally bound peptide ligands. Importantly, CTL activated in this manner can break self-tolerance by reacting to self-peptides presented by native MHC. The ability to activate self-tolerant T cells capable of cross-reacting on self-peptide-MHC in vivo represents an approach for inducing autoimmunity, with possible application in cancer vaccines.


Assuntos
Apresentação de Antígeno/imunologia , Citotoxicidade Imunológica , Antígenos de Histocompatibilidade Classe I/imunologia , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos/genética , Animais , Linfócitos T CD8-Positivos/imunologia , Humanos , Tolerância Imunológica , Ligantes , Ativação Linfocitária/imunologia , Camundongos , Peptídeos/genética , Peptídeos/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
7.
Blood ; 133(7): 697-709, 2019 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-30463995

RESUMO

Chimeric antigen receptor T (CAR-T) cell therapy is a new pillar in cancer therapeutics; however, its application is limited by the associated toxicities. These include cytokine release syndrome (CRS) and neurotoxicity. Although the IL-6R antagonist tocilizumab is approved for treatment of CRS, there is no approved treatment of neurotoxicity associated with CD19-targeted CAR-T (CART19) cell therapy. Recent data suggest that monocytes and macrophages contribute to the development of CRS and neurotoxicity after CAR-T cell therapy. Therefore, we investigated neutralizing granulocyte-macrophage colony-stimulating factor (GM-CSF) as a potential strategy to manage CART19 cell-associated toxicities. In this study, we show that GM-CSF neutralization with lenzilumab does not inhibit CART19 cell function in vitro or in vivo. Moreover, CART19 cell proliferation was enhanced and durable control of leukemic disease was maintained better in patient-derived xenografts after GM-CSF neutralization with lenzilumab. In a patient acute lymphoblastic leukemia xenograft model of CRS and neuroinflammation (NI), GM-CSF neutralization resulted in a reduction of myeloid and T cell infiltration in the central nervous system and a significant reduction in NI and prevention of CRS. Finally, we generated GM-CSF-deficient CART19 cells through CRISPR/Cas9 disruption of GM-CSF during CAR-T cell manufacturing. These GM-CSFk/o CAR-T cells maintained normal functions and had enhanced antitumor activity in vivo, as well as improved overall survival, compared with CART19 cells. Together, these studies illuminate a novel approach to abrogate NI and CRS through GM-CSF neutralization, which may potentially enhance CAR-T cell function. Phase 2 studies with lenzilumab in combination with CART19 cell therapy are planned.


Assuntos
Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Doenças do Sistema Imunitário/terapia , Inflamação/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfócitos T/uso terapêutico , Receptores de Antígenos Quiméricos/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Proliferação de Células , Humanos , Doenças do Sistema Imunitário/imunologia , Doenças do Sistema Imunitário/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Síndrome , Transplante Heterólogo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Brain ; 143(12): 3629-3652, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33253355

RESUMO

Immunosuppression of unknown aetiology is a hallmark feature of glioblastoma and is characterized by decreased CD4 T-cell counts and downregulation of major histocompatibility complex class II expression on peripheral blood monocytes in patients. This immunosuppression is a critical barrier to the successful development of immunotherapies for glioblastoma. We recapitulated the immunosuppression observed in glioblastoma patients in the C57BL/6 mouse and investigated the aetiology of low CD4 T-cell counts. We determined that thymic involution was a hallmark feature of immunosuppression in three distinct models of brain cancer, including mice harbouring GL261 glioma, B16 melanoma, and in a spontaneous model of diffuse intrinsic pontine glioma. In addition to thymic involution, we determined that tumour growth in the brain induced significant splenic involution, reductions in peripheral T cells, reduced MHC II expression on blood leucocytes, and a modest increase in bone marrow resident CD4 T cells. Using parabiosis we report that thymic involution, declines in peripheral T-cell counts, and reduced major histocompatibility complex class II expression levels were mediated through circulating blood-derived factors. Conversely, T-cell sequestration in the bone marrow was not governed through circulating factors. Serum isolated from glioma-bearing mice potently inhibited proliferation and functions of T cells both in vitro and in vivo. Interestingly, the factor responsible for immunosuppression in serum is non-steroidal and of high molecular weight. Through further analysis of neurological disease models, we determined that the immunosuppression was not unique to cancer itself, but rather occurs in response to brain injury. Non-cancerous acute neurological insults also induced significant thymic involution and rendered serum immunosuppressive. Both thymic involution and serum-derived immunosuppression were reversible upon clearance of brain insults. These findings demonstrate that brain cancers cause multifaceted immunosuppression and pinpoint circulating factors as a target of intervention to restore immunity.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Tolerância Imunológica , Mediadores da Inflamação/metabolismo , Animais , Células da Medula Óssea/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células , Progressão da Doença , Feminino , Genes MHC da Classe II/genética , Glioblastoma/imunologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioma/imunologia , Glioma/metabolismo , Glioma/patologia , Masculino , Melanoma Experimental/imunologia , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Parabiose , Convulsões/induzido quimicamente , Baço/imunologia , Baço/patologia , Theilovirus , Timo/patologia
9.
Liver Transpl ; 26(4): 549-563, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31950576

RESUMO

The liver is an immunologically active organ with a tolerogenic microenvironment at a quiescent state. The immunoregulatory properties of the liver appear to be retained after transplantation because liver allografts can reduce alloresponses against other organs that are simultaneously transplanted. Mechanisms of this phenomenon remain unknown. Given the known immunomodulatory properties of mesenchymal stromal cells (MSCs), we hypothesized that liver mesenchymal stromal cells (L-MSCs) are superior immunomodulators and contribute to liver-mediated tolerance. L-MSCs, generated from human liver allograft biopsies, were compared with adipose mesenchymal stromal cells (A-MSCs) and bone marrow mesenchymal stromal cells (BM-MSCs). Trilineage differentiation of L-MSCs was confirmed by immunohistochemistry. Comparative phenotypic analyses were done by flow cytometry and transcriptome analyses by RNA sequencing in unaltered cell cultures. The in vitro functional analyses were performed using alloreactive T cell proliferation assays. The transcriptome analysis showed that the L-MSCs are different than the A-MSCs and BM-MSCs, with significant enrichment of genes and gene sets associated with immunoregulation. Compared with the others, L-MSCs were found to express higher cell surface levels of several select immunomodulatory molecules. L-MSCs (versus A-MSCs/BM-MSCs) inhibited alloreactive T cell proliferation (22.7% versus 56.4%/58.7%, respectively; P < 0.05) and reduced the frequency of interferon ɤ-producing T cells better than other MSCs (52.8% versus 94.4%/155.4%; P < 0.05). The antiproliferative impact of L-MSCs was not dependent on cell-to-cell contact, could be reversed incompletely by blocking programmed death ligand 1, and required a higher concentration of the competitive inhibitor of indoleamine 2,3-dioxygenase for complete reversal. In conclusion, L-MSCs appear to be uniquely well-equipped immunomodulatory cells, and they are more potent than A-MSCs and BM-MSCs in that capacity, which suggests that they may contribute to liver-induced systemic tolerance.


Assuntos
Transplante de Fígado , Células-Tronco Mesenquimais , Células da Medula Óssea , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos , Imunomodulação , Fígado
10.
Environ Sci Technol ; 54(12): 7639-7650, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32407626

RESUMO

Gaseous emissions from livestock production are complex mixtures including ammonia, methane, volatile organic compounds (VOC), and H2S. These contribute to eutrophication, reduced air quality, global warming, and odor nuisance. It is imperative that these gases are mitigated in an environmentally sustainable manner. We present the discovery of a microbial inhibitor combo consisting of tannic acid and sodium fluoride (TA-NaF), which exhibits clear synergistic inhibition of ammonia production in pure bacteria culture and in pig manure while simultaneously inhibiting methane and odorant (H2S and VOC) emissions. In laboratory headspace experiments on pig manure, we used proton-transfer-reaction mass spectrometry and cavity ring-down spectroscopy to measure the effect of TA-NaF on gaseous emissions. Ammonia emission was reduced by more than 95%, methane by up to ∼99%, and odor activity value by more than 50%. Microbial community analysis and gas emission data suggest that TA-NaF acts as an efficient generic microbial inhibitor, and we hypothesize that the synergistic inhibitory effect on ammonia production is related to tannic acid causing cell membrane leakage allowing fluoride ions easy access to urease.


Assuntos
Amônia , Metano , Amônia/análise , Animais , Fluoretos , Gado , Esterco , Odorantes , Suínos , Taninos
11.
Kidney Int ; 93(6): 1465-1474, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29656904

RESUMO

Kidney allografts of patients who undergo simultaneous liver-kidney transplantation incur less immune-mediated injury, and retain better function compared to other kidney allografts. To characterize the host alloimmune responses in 28 of these patients, we measured the donor-specific alloresponsiveness and phenotypes of peripheral blood cells after the first year. These values were then compared to those of 61 similarly immunosuppressed recipients of a solitary kidney or 31 recipients of liver allografts. Four multicolor, non-overlapping flow cytometry protocols were used to assess the immunophenotypes. Mixed cell cultures with donor or third party cells were used to measure cell proliferation and interferon gamma production. Despite a significant overlap, simultaneous liver-kidney transplant recipients had a lower overall frequency of circulating CD8+, activated CD4+ and effector memory T cells, compared to solitary kidney transplant recipients. Simultaneous liver-kidney transplant recipient T cells had a significantly lower proliferative response to the donor cells compared to solitary kidney recipients (11.9 vs. 42.9%), although their response to third party cells was unaltered. The frequency of interferon gamma producing alloreactive T cells in simultaneous liver-kidney transplant recipients was significantly lower than that of solitary kidney transplant recipients. Flow cytometric analysis of the mixed cultures demonstrated that both alloreactive CD4+ and CD8+ compartments of the simultaneous liver-kidney transplant recipient circulating blood cells were smaller. Thus, the phenotypic and functional characteristics of the circulating blood cells of the simultaneous liver-kidney transplant recipients resembled those of solitary liver transplant recipients, and appear to be associated with donor-specific hypo-alloresponsiveness.


Assuntos
Antígenos HLA-A/imunologia , Histocompatibilidade , Isoanticorpos/imunologia , Transplante de Rim , Transplante de Fígado , Ativação Linfocitária , Subpopulações de Linfócitos T/imunologia , Adulto , Idoso , Aloenxertos , Biomarcadores/sangue , Células Cultivadas , Feminino , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto , Humanos , Imunossupressores/uso terapêutico , Isoanticorpos/sangue , Transplante de Rim/efeitos adversos , Transplante de Fígado/efeitos adversos , Masculino , Pessoa de Meia-Idade , Fenótipo , Fatores de Risco , Subpopulações de Linfócitos T/metabolismo , Fatores de Tempo , Resultado do Tratamento
12.
Environ Sci Technol ; 52(4): 2100-2107, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29338206

RESUMO

Reduced sulfur compounds emitted from livestock production cause odor nuisance for local residents. The microbial processes responsible for this are not well described in swine manure and a method for monitoring the biological processes is necessary to develop strategic abatement technologies. In this study, Proton-Transfer-Reaction Mass Spectrometry and isotope-labeled sulfate were combined and applied to elucidate the sulfur processes in swine manure with high time resolution. We successfully monitored reduction of isotope 33S labeled sulfate into corresponding 33S hydrogen sulfide and found that some of the 33S hydrogen sulfide was further methylated into 33S methanethiol. The isotope patterns in reduced sulfur compounds together with usage of inhibitors enabled us to calculate a sulfate reduction rate of 1.03 ± 0.18 mM/day equivalent to 76.9 ± 3.0% of total hydrogen sulfide emissions. Cysteine degradation constituted 20.2 ± 2.7% of the total hydrogen sulfide produced and the remaining hydrogen sulfide came from demethylation of methanethiol and dimethyl sulfide. Another source to methanethiol, besides hydrogen sulfide methylation, was methionine degradation, which contributed with 78.3 ± 2.5% of the methanethiol production, whereas the remaining 21.7 ± 2.5% came from hydrogen sulfide methylation. This study suggests, therefore, that emissions of odorous sulfur compounds from swine manure can be reduced by inhibiting methionine degradation and sulfate reduction.


Assuntos
Gado , Enxofre , Animais , Marcação por Isótopo , Espectrometria de Massas , Odorantes , Prótons , Suínos
13.
Water Sci Technol ; 77(5-6): 1751-1756, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29595178

RESUMO

Odor analysis by olfactometry relies on the use of n-butanol as a reference compound for standardizing the selection of human panelists. This requires that human sensitivity towards n-butanol is correlated to sensitivity towards other odorants as well as complex odor mixtures. However, there is limited evidence in the literature of such correlations. In this work, datasets from three odor laboratories were investigated in order to clarify this. All panels routinely analyzed n-butanol and H2S samples. Two of the laboratories analyzed samples from pig production or industry, whereas one laboratory determined odor threshold values for typical pig production odorants. Non-significant correlations were observed in most cases and odor threshold values for structurally related compounds were not well correlated. The work presented strongly indicates that the sensitivity of odor panelists towards n-butanol is not well transferred to other odorants or odor samples. Furthermore, minimization of variance by using n-butanol is not transferable to other odorants or environmental samples. Thus, the harmonization of human panelists for odor analysis based on n-butanol does not appear to result in harmonization with respect to other odorants or odor samples.


Assuntos
1-Butanol/química , Odorantes/análise , Poluentes Atmosféricos , Animais , Monitoramento Ambiental , Humanos , Olfatometria , Valores de Referência , Suínos
14.
J Immunol ; 195(12): 5648-56, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26561552

RESUMO

Calcium-modulating cyclophilin ligand (CAML) is an endoplasmic reticulum resident protein that is widely expressed. Although it has been demonstrated to participate in the tail-anchored protein insertion pathway, its physiological role in the mature immune system is unknown. In this work, we show that mature, peripheral T cells require CAML for survival specifically following TCR-induced activation. In this study, we examined mature T cells from spleen and lymph nodes of tamoxifen-inducible CAML knockout mice (tCAML(-/-)). Whereas CAML-deficient T cells were able to express the early activation markers CD25 and CD69, and produce IL-2 normally upon stimulation, deficient cells proliferated less and died. Cells did not require CAML for entry into the S phase of the cell cycle, thus implicating its survival function at a relatively late step in the T cell activation sequence. In addition, CAML was required for homeostatic proliferation and for Ag-dependent cell killing in vivo. These results demonstrate that CAML critically supports T cell survival and cell division downstream of T cell activation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cálcio/metabolismo , Ciclofilinas/metabolismo , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Imunidade Adaptativa , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Geneticamente Modificados , Sobrevivência Celular , Células Cultivadas , Ligantes , Ativação Linfocitária , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia
15.
BMC Evol Biol ; 16(1): 219, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27756206

RESUMO

BACKGROUND: Adaptive radiation involving a colonizing phenotype that rapidly evolves into at least one other ecological variant, or ecotype, has been observed in a variety of freshwater fishes in post-glacial environments. However, few studies consider how phenotypic traits vary with regard to neutral genetic partitioning along ecological gradients. Here, we present the first detailed investigation of lake trout Salvelinus namaycush that considers variation as a cline rather than discriminatory among ecotypes. Genetic and phenotypic traits organized along common ecological gradients of water depth and geographic distance provide important insights into diversification processes in a lake with high levels of human disturbance from over-fishing. RESULTS: Four putative lake trout ecotypes could not be distinguished using population genetic methods, despite morphological differences. Neutral genetic partitioning in lake trout was stronger along a gradient of water depth, than by locality or ecotype. Contemporary genetic migration patterns were consistent with isolation-by-depth. Historical gene flow patterns indicated colonization from shallow to deep water. Comparison of phenotypic (Pst) and neutral genetic variation (Fst) revealed that morphological traits related to swimming performance (e.g., buoyancy, pelvic fin length) departed more strongly from neutral expectations along a depth gradient than craniofacial feeding traits. Elevated phenotypic variance with increasing water depth in pelvic fin length indicated possible ongoing character release and diversification. Finally, differences in early growth rate and asymptotic fish length across depth strata may be associated with limiting factors attributable to cold deep-water environments. CONCLUSION: We provide evidence of reductions in gene flow and divergent natural selection associated with water depth in Lake Superior. Such information is relevant for documenting intraspecific biodiversity in the largest freshwater lake in the world for a species that recently lost considerable genetic diversity and is now in recovery. Unknown is whether observed patterns are a result of an early stage of incipient speciation, gene flow-selection equilibrium, or reverse speciation causing formerly divergent ecotypes to collapse into a single gene pool.


Assuntos
Ecossistema , Variação Genética , Lagos , Truta/genética , Animais , Fluxo Gênico , Genética Populacional , Geografia , Fenótipo , Densidade Demográfica , Seleção Genética , Água
17.
Inflamm Res ; 64(9): 697-706, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26149693

RESUMO

OBJECTIVE AND DESIGN: Adipose tissue macrophages (ATMs) have been implicated in a number of obesity-related diseases. Because the activated macrophages associated with many types of autoimmune and inflammatory diseases express a folate receptor (FR) that can be exploited for FR-targeted drug delivery, we examined the visceral adipose tissue of obese mice and humans to determine whether ATMs also express FR that are accessible by folate conjugates. MATERIAL OR SUBJECTS: C57BL/6 or FATSO mice fed on either a low- or high-fat diet were used in murine studies. Human adipose tissue were obtained from healthy volunteers during adipose reduction surgery. METHODS: Visceral adipose tissue was collected from both obese mice and humans, collagenase digested, and stained with folate-Oregon Green and antibodies for macrophage markers including F4/80, mannose receptor (CD206), CD11b, and CD11c. Cells were then examined for expression of the above markers by flow cytometry. Furthermore, the ability of folate conjugates to target the FR-expressing ATMs in obese mice was evaluated in vivo. RESULTS: A subset of the ATMs harvested from obese mice were found to express FR. Subpopulations of ATMs also simultaneously express both pro- and anti-inflammatory markers, and FR is expressed on both subsets. We then demonstrate that FR-expressing ATMs can be targeted with folate-linked fluorescent dyes in vivo. CONCLUSIONS: FR are expressed on multiple subsets of ATMs and these subsets can be targeted with folate-linked drugs, allowing for the possible development of FR-targeted therapies for obesity-related inflammatory diseases.


Assuntos
Tecido Adiposo/citologia , Receptores de Folato com Âncoras de GPI/metabolismo , Macrófagos/metabolismo , Obesidade/metabolismo , Animais , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Dieta Hiperlipídica , Receptores de Folato com Âncoras de GPI/imunologia , Humanos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/imunologia
18.
PLoS Pathog ; 8(2): e1002541, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22383876

RESUMO

Structural diversity in the peptide binding sites of the redundant classical MHC antigen presenting molecules is strongly selected in humans and mice. Although the encoded antigen presenting molecules overlap in antigen presenting function, differences in polymorphism at the MHC I A, B and C loci in humans and higher primates indicate these loci are not functionally equivalent. The structural basis of these differences is not known. We hypothesize that classical class I loci differ in their ability to direct effective immunity against intracellular pathogens. Using a picornavirus infection model and chimeric H-2 transgenes, we examined locus specific functional determinants distinguishing the ability of class I sister genes to direct effective anti viral immunity. Whereas, parental FVB and transgenic FVB mice expressing the H-2K(b) gene are highly susceptible to persisting Theiler's virus infection within the CNS and subsequent demyelination, mice expressing the D(b) transgene clear the virus and are protected from demyelination. Remarkably, animals expressing a chimeric transgene, comprised primarily of K(b) but encoding the peptide binding domain of D(b), develop a robust anti viral CTL response yet fail to clear virus and develop significant demyelination. Differences in expression of the chimeric K(b)α1α2D(b) gene (low) and D(b) (high) in the CNS of infected mice mirror expression levels of their endogenous H-2(q) counterparts in FVB mice. These findings demonstrate that locus specific elements other than those specifying peptide binding and T cell receptor interaction can determine ability to clear virus infection. This finding provides a basis for understanding locus-specific differences in MHC polymorphism, characterized best in human populations.


Assuntos
Genes MHC Classe I/fisiologia , Loci Gênicos/fisiologia , Imunidade Inata/genética , Vírus/imunologia , Animais , Eficiência , Antígenos H-2/química , Antígenos H-2/genética , Antígenos H-2/metabolismo , Células HEK293 , Antígeno de Histocompatibilidade H-2D , Humanos , Imunidade Inata/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Transgênicos , Modelos Moleculares , Viroses/genética , Viroses/imunologia
19.
Mol Pharm ; 11(3): 1007-13, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24446917

RESUMO

The folate receptor (FR) is a GPI anchored cell surface glycoprotein that functions to facilitate folic acid uptake and mediate signal transduction. With the introduction of multiple folate-targeted drugs into the clinic, the question has arisen regarding how frequently a patient can be dosed with a FR-targeted drug or antibody and whether dosing frequency exerts any impact on the availability of FR for subsequent rounds of FR-mediated drug uptake. Although the rate of FR recycling has been examined in murine tumor models, little or no information exists on the impact of FR occupancy on its rate of endocytosis. The present study quantitates the number of cell surface FR-α and FR-ß following exposure to saturating concentrations of a variety of folate-linked molecules and anti-FR antibodies, including the unmodified vitamin, folate-linked drug mimetics, multifolate derivatized nanoparticles, and monoclonal antibodies to FR. We report here that FR occupancy has no impact on the rate of FR internalization. We also demonstrate that multivalent conjugates that bind and cross-link FRs at the cell surface internalize at the same rate as monovalent folate conjugates that have no impact on FR clustering, even though the multivalent conjugates traffic through a different endocytic pathway.


Assuntos
Anticorpos Monoclonais/metabolismo , Endocitose/fisiologia , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Ácido Fólico/metabolismo , Neoplasias Bucais/metabolismo , Neoplasias Ovarianas/metabolismo , Membrana Celular/metabolismo , Feminino , Humanos , Células Tumorais Cultivadas
20.
Mol Ther ; 21(5): 1087-95, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23568262

RESUMO

Picornaviruses have been developed as potential therapies for gene delivery and vaccination. One drawback to their use is the potential for recombination and viral persistence. Therefore, the engineering strategies used must take into account the possibility for virus escape. We have developed Theiler's murine encephalomyelitis virus (TMEV) as a potential vaccine vector for use in immunotherapy. This study shows that insertion of a vaccine epitope at a unique site within the TMEV leader protein can dramatically increase the type I interferon (IFN) response to infection and promote rapid viral clearance. This live virus vaccine maintains its ability to drive antigen-specific CD8(+) T-cell responses to a model antigen as well as to the weakly immunogenic tumor antigen Her2/neu. Furthermore, the epitope integration site does not affect the efficacy of this vaccine as cancer immunotherapy for treating models of melanoma and breast cancer as demonstrated by delayed tumor outgrowth and increased survival in animals implanted with these tumors. These findings show that an attenuated virus retaining limited ability to replicate nonetheless can effectively mobilize CD8(+) cellular immunity and will be important for the design of picornavirus vectors used as immunotherapy in clinical settings.


Assuntos
Antígenos/imunologia , Vacinas Anticâncer/imunologia , Epitopos/imunologia , Neoplasias/imunologia , Theilovirus/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/mortalidade , Infecções por Cardiovirus/virologia , Linhagem Celular Tumoral , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Imunoterapia , Interferon Tipo I/imunologia , Camundongos , Dados de Sequência Molecular , Mutagênese Insercional , Neoplasias/patologia , Neoplasias/terapia , Receptor ErbB-2/imunologia , Theilovirus/genética , Carga Tumoral/efeitos dos fármacos , Vacinas Atenuadas , Proteínas Virais/química , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa