Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Theranostics ; 14(4): 1647-1661, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38389838

RESUMO

Background: Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods: BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results: BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion: When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.


Assuntos
Ablação por Ultrassom Focalizado de Alta Intensidade , Melanoma , Humanos , Melanoma/terapia , Ablação por Ultrassom Focalizado de Alta Intensidade/métodos , Antígenos de Neoplasias , Células Dendríticas
2.
bioRxiv ; 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-37732205

RESUMO

Background: Boiling histotripsy (BH), a mechanical focused ultrasound ablation strategy, can elicit intriguing signatures of anti-tumor immunity. However, the influence of BH on dendritic cell function is unknown, compromising our ability to optimally combine BH with immunotherapies to control metastatic disease. Methods: BH was applied using a sparse scan (1 mm spacing between sonications) protocol to B16F10-ZsGreen melanoma in bilateral and unilateral settings. Ipsilateral and contralateral tumor growth was measured. Flow cytometry was used to track ZsGreen antigen and assess how BH drives dendritic cell behavior. Results: BH monotherapy elicited ipsilateral and abscopal tumor control in this highly aggressive model. Tumor antigen presence in immune cells in the tumor-draining lymph nodes (TDLNs) was ~3-fold greater at 24h after BH, but this abated by 96h. B cells, macrophages, monocytes, granulocytes, and both conventional dendritic cell subsets (i.e. cDC1s and cDC2s) acquired markedly more antigen with BH. BH drove activation of both cDC subsets, with activation being dependent upon tumor antigen acquisition. Our data also suggest that BH-liberated tumor antigen is complexed with damage-associated molecular patterns (DAMPs) and that cDCs do not traffic to the TDLN with antigen. Rather, they acquire antigen as it flows through afferent lymph vessels into the TDLN. Conclusion: When applied with a sparse scan protocol, BH monotherapy elicits abscopal melanoma control and shapes dendritic cell function through several previously unappreciated mechanisms. These results offer new insight into how to best combine BH with immunotherapies for the treatment of metastatic melanoma.

4.
Blood ; 136(18): 2065-2079, 2020 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-32556153

RESUMO

Macrophage antibody (Ab)-dependent cellular phagocytosis (ADCP) is a major cytotoxic mechanism for both therapeutic unconjugated monoclonal Abs (mAbs) such as rituximab and Ab-induced hemolytic anemia and immune thrombocytopenia. Here, we studied the mechanisms controlling the rate and capacity of macrophages to carry out ADCP in settings of high target/effector cell ratios, such as those seen in patients with circulating tumor burden in leukemic phase disease. Using quantitative live-cell imaging of primary human and mouse macrophages, we found that, upon initial challenge with mAb-opsonized lymphocytes, macrophages underwent a brief burst (<1 hour) of rapid phagocytosis, which was then invariably followed by a sharp reduction in phagocytic activity that could persist for days. This previously unknown refractory period of ADCP, or hypophagia, was observed in all macrophage, mAb, and target cell conditions tested in vitro and was also seen in vivo in Kupffer cells from mice induced to undergo successive rounds of αCD20 mAb-dependent clearance of circulating B cells. Importantly, hypophagia had no effect on Ab-independent phagocytosis and did not alter macrophage viability. In mechanistic studies, we found that the rapid loss of activating Fc receptors from the surface and their subsequent proteolytic degradation were the primary mechanisms responsible for the loss of ADCP activity in hypophagia. These data suggest hypophagia is a critical limiting step in macrophage-mediated clearance of cells via ADCP, and understanding such limitations to innate immune system cytotoxic capacity will aid in the development of mAb regimens that could optimize ADCP and improve patient outcome.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Imunidade Inata/efeitos dos fármacos , Macrófagos/patologia , Fagócitos/imunologia , Fagocitose , Rituximab/farmacologia , Animais , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagócitos/efeitos dos fármacos
5.
J Cell Sci ; 133(5)2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32005699

RESUMO

Phagocytosis is a dynamic process central to immunity and tissue homeostasis. Current methods for quantification of phagocytosis largely rely on indirect or static measurements, such as target clearance or dye uptake, and thus provide limited information about engulfment rates or target processing. Improved kinetic measurements of phagocytosis could provide useful, basic insights in many areas. We present a live-cell, time-lapse and high-content microscopy imaging method based on the detection and quantification of fluorescent dye 'voids' within phagocytes that result from target internalization to quantify phagocytic events with high temporal resolution. Using this method, we measure target cell densities and antibody concentrations needed for optimal antibody-dependent cellular phagocytosis. We compare void formation and dye uptake methods for phagocytosis detection, and examine the connection between target cell engulfment and phagolysosomal processing. We demonstrate how this approach can be used to measure distinct forms of phagocytosis, and changes in macrophage morphology during phagocytosis related to both engulfment and target degradation. Our results provide a high-resolution method for quantifying phagocytosis that provides opportunities to better understand the cellular and molecular regulation of this fundamental biological process.


Assuntos
Microscopia , Fagócitos , Macrófagos , Fagocitose , Imagem com Lapso de Tempo
6.
J Neuroinflammation ; 16(1): 261, 2019 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-31822279

RESUMO

BACKGROUND: Neuroinflammation is thought to contribute to the pathogenesis of Alzheimer's disease (AD), yet numerous studies have demonstrated a beneficial role for neuroinflammation in amyloid plaque clearance. We have previously shown that sustained expression of IL-1ß in the hippocampus of APP/PS1 mice decreases amyloid plaque burden independent of recruited CCR2+ myeloid cells, suggesting resident microglia as the main phagocytic effectors of IL-1ß-induced plaque clearance. To date, however, the mechanisms of IL-1ß-induced plaque clearance remain poorly understood. METHODS: To determine whether microglia are involved in IL-1ß-induced plaque clearance, APP/PS1 mice induced to express mature human IL-1ß in the hippocampus via adenoviral transduction were treated with the Aß fluorescent probe methoxy-X04 (MX04) and microglial internalization of fibrillar Aß (fAß) was analyzed by flow cytometry and immunohistochemistry. To assess microglial proliferation, APP/PS1 mice transduced with IL-1ß or control were injected intraperitoneally with BrdU and hippocampal tissue was analyzed by flow cytometry. RNAseq analysis was conducted on microglia FACS sorted from the hippocampus of control or IL-1ß-treated APP/PS1 mice. These microglia were also sorted based on MX04 labeling (MX04+ and MX04- microglia). RESULTS: Resident microglia (CD45loCD11b+) constituted > 70% of the MX04+ cells in both Phe- and IL-1ß-treated conditions, and < 15% of MX04+ cells were recruited myeloid cells (CD45hiCD11b+). However, IL-1ß treatment did not augment the percentage of MX04+ microglia nor the quantity of fAß internalized by individual microglia. Instead, IL-1ß increased the total number of MX04+ microglia in the hippocampus due to IL-1ß-induced proliferation. In addition, transcriptomic analyses revealed that IL-1ß treatment was associated with large-scale changes in the expression of genes related to immune responses, proliferation, and cytokine signaling. CONCLUSIONS: These studies show that IL-1ß overexpression early in amyloid pathogenesis induces a change in the microglial gene expression profile and an expansion of microglial cells that facilitates Aß plaque clearance.


Assuntos
Reprogramação Celular/fisiologia , Interleucina-1beta/biossíntese , Microglia/metabolismo , Placa Amiloide/metabolismo , Transcrição Gênica/fisiologia , Transcriptoma/fisiologia , Animais , Proliferação de Células/fisiologia , Feminino , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Placa Amiloide/genética
7.
JCI Insight ; 52019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30998506

RESUMO

The bone marrow microenvironment (BMME) contributes to the regulation of hematopoietic stem cell (HSC) function, though its role in age-associated lineage skewing is poorly understood. Here we show that dysfunction of aged marrow macrophages (Mφs) directs HSC platelet-bias. Mφs from the marrow of aged mice and humans exhibited an activated phenotype, with increased expression of inflammatory signals. Aged marrow Mφs also displayed decreased phagocytic function. Senescent neutrophils, typically cleared by marrow Mφs, were markedly increased in aged mice, consistent with functional defects in Mφ phagocytosis and efferocytosis. In aged mice, Interleukin 1B (IL1B) was elevated in the bone marrow and caspase 1 activity, which can process pro-IL1B, was increased in marrow Mφs and neutrophils. Mechanistically, IL1B signaling was necessary and sufficient to induce a platelet bias in HSCs. In young mice, depletion of phagocytic cell populations or loss of the efferocytic receptor Axl expanded platelet-biased HSCs. Our data support a model wherein increased inflammatory signals and decreased phagocytic function of aged marrow Mφs induce the acquisition of platelet bias in aged HSCs. This work highlights the instructive role of Mφs and IL1B in the age-associated lineage-skewing of HSCs, and reveals the therapeutic potential of their manipulation as antigeronic targets.


Assuntos
Envelhecimento/fisiologia , Plaquetas/metabolismo , Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Animais , Medula Óssea/patologia , Caspase 1/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos , Fagocitose , Fenótipo , Proteínas Proto-Oncogênicas , Receptores Proteína Tirosina Quinases , Receptor Tirosina Quinase Axl
8.
Front Immunol ; 9: 2131, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30283457

RESUMO

Invasive fungal infections, including Pneumocystis Pneumonia (PcP), remain frequent life-threatening conditions of patients with adaptive immune defects. While innate immunity helps control pathogen growth early during infection, it is typically not sufficient for complete protection against Pneumocystis and other human fungal pathogens. Alveolar macrophages (AM) possess pattern recognition molecules capable of recognizing antigenic and structural determinants of Pneumocystis. However, this pathogen effectively evades innate immunity to infect both immunocompetent and immunosuppressed hosts, albeit with differing outcomes. During our studies of mouse models of PcP, the FVB/N strain was identified as unique because of its ability to mount a protective innate immune response against Pneumocystis infection. In contrast to other immunocompetent strains, which become transiently infected prior to the onset of adaptive immunity, FVB/N mice rapidly eradicated Pneumocystis before an adaptive immune response was triggered. Furthermore, FVB/N mice remained highly resistant to infection even in the absence of functional T cells. The effector mechanism of innate protection required the action of functional alveolar macrophages, and the adoptive transfer of resistant FVB/N AMs, but not susceptible CB.17 AMs, conferred protection to immunodeficient mice. Macrophage IFNγ receptor signaling was not required for innate resistance, and FVB/N macrophages were found to display markers of alternative activation. IFNγ reprogrammed resistant FVB/N macrophages to a permissive M1 biased phenotype through a mechanism that required direct activation of the macrophage IFNγR. These results demonstrate that appropriately programmed macrophages provide protective innate immunity against this opportunistic fungal pathogen, and suggest that modulating macrophage function may represent a feasible therapeutic strategy to enhance antifungal host defense. The identification of resistant and susceptible macrophages provides a novel platform to study not only the mechanisms of macrophage-mediated antifungal defense, but also the mechanisms by which Pneumocystis evades innate immunity.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Macrófagos Alveolares/imunologia , Infecções por Pneumocystis/imunologia , Animais , Humanos , Imunocompetência , Hospedeiro Imunocomprometido , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos Alveolares/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Infecções por Pneumocystis/microbiologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
9.
Neurobiol Dis ; 69: 124-33, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24874542

RESUMO

Neuroinflammation is a key component of Alzheimer's disease (AD) pathogenesis. Particularly, the proinflammatory cytokine interleukin-1 beta (IL-1ß) is upregulated in human AD and believed to promote amyloid plaque deposition. However, studies from our laboratory have shown that chronic IL-1ß overexpression in the APPswe/PSEN1dE9 (APP/PS1) mouse model of AD ameliorates amyloid pathology, increases plaque-associated microglia, and induces recruitment of peripheral immune cells to the brain parenchyma. To investigate the contribution of CCR2 signaling in IL-1ß-mediated amyloid plaque clearance, seven month-old APP/PS1/CCR2(-/-) mice were intrahippocampally transduced with a recombinant adeno-associated virus serotype 2 containing the cleaved form of human IL-1ß (rAAV2-IL-1ß). Four weeks after rAAV2-IL-1ß transduction, we found significant reductions in 6E10 and Congo red staining of amyloid plaques that was confirmed by decreased levels of insoluble Aß1-42 and Aß1-40 in the inflamed hippocampus. Bone marrow chimeric studies confirmed the presence of infiltrating immune cells following IL-1ß overexpression and revealed that dramatic reduction of CCR2(+) peripheral mononuclear cell recruitment to the inflamed hippocampus did not prevent the ability of IL-1ß to induce amyloid plaque clearance. These results suggest that infiltrating CCR2(+) monocytes do not contribute to IL-1ß-mediated amyloid plaque clearance.


Assuntos
Doença de Alzheimer/imunologia , Hipocampo/imunologia , Interleucina-1beta/metabolismo , Placa Amiloide/imunologia , Receptores CCR2/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Hipocampo/patologia , Humanos , Interleucina-1beta/genética , Leucócitos Mononucleares/imunologia , Camundongos Transgênicos , Neuroimunomodulação/fisiologia , Fragmentos de Peptídeos/metabolismo , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores CCR2/genética , Transdução de Sinais , Quimeras de Transplante
10.
Brain Behav Immun ; 32: 9-18, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23510988

RESUMO

Alterations in adult hippocampal neurogenesis have been observed in numerous neurological diseases that contain a neuroinflammatory component. Interleukin-1ß (IL-1ß) is a pro-inflammatory cytokine that contributes to neuroinflammation in many CNS disorders. Our previous results reveal a severe reduction in adult hippocampal neurogenesis due to focal and chronic expression of IL-1ß in a transgenic mouse model, IL-1ß(XAT), that evokes a complex neuroinflammatory response. Other investigators have shown that IL-1ß can bind directly to neural precursors to cause cell cycle arrest in vitro. In order to observe if IL-1 signaling is necessary in vivo, we conditionally knocked out MyD88, an adapter protein essential for IL-1 signaling, in nestin(+) neural precursor cells (NPCs) in the presence of IL-1ß-dependent inflammation. Our results show that conditional knockout of MyD88 does not prevent IL-1ß-induced reduction in neuroblasts using a genetic fate mapping model. Interestingly, MyD88 deficiency in nestin(+) NPCs causes an increase in the number of astrocytes in the presence of IL-1ß, suggesting that MyD88-dependent signaling is important in limiting astroglial differentiation due to inflammation. MyD88 deficiency does not alter the fate of NPCs in the absence of inflammation. Furthermore, the inflammatory milieu due to IL-1ß is not affected by the absence of MyD88 in nestin(+) NPCs. These results show that sustained IL-1ß causes a reduction in adult hippocampal neurogenesis that is independent of MyD88-dependent signaling in nestin(+) NPCs, suggesting an indirect negative effect of IL-1ß on neurogenesis.


Assuntos
Hipocampo/crescimento & desenvolvimento , Interleucina-1beta/biossíntese , Nestina/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Antineoplásicos Hormonais/toxicidade , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Dependovirus/genética , Feminino , Citometria de Fluxo , Hipocampo/citologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/biossíntese , Fator 88 de Diferenciação Mieloide/genética , Nestina/genética , Células-Tronco Neurais/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina-1/biossíntese , Transdução de Sinais/fisiologia , Tamoxifeno/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...