Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(33): 16513-16518, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31363052

RESUMO

Efferocytosis of apoptotic neutrophils (PMNs) by alveolar macrophages (AMФs) is vital for resolution of inflammation and tissue injury. Here, we investigated the role of AMФ polarization and expression of the efferocytic ligand Gas6 in restoring homeostasis. In the murine model of lipopolysaccharide (LPS)-induced acute lung injury (ALI), we observed augmented temporal generation of cytokines IL-4 and TSG6 in bronchoalveolar fluid (BALF). Interestingly, we also observed increased expression of antiinflammatory markers consistent with a phenotype shift in AMФs. In particular, AMФs expressed the efferocytic ligand Gas6. In vitro priming of bone marrow-derived macrophages (BMMФs) with IL-4 or TSG6 also induced MФ transition and expression of Gas6. TSG6- or IL-4-primed BMMФs induced efferocytosis of apoptotic PMNs compared with control BMMФs. Adoptive transfer of TSG6- or IL-4-primed BMMФs i.t. into LPS-challenged mice more rapidly and effectively cleared PMNs in lungs compared with control BMMФs. We demonstrated that expression of Gas6 during AMФ transition was due to activation of the transcription factor signal transducer and activator of transcription-6 (STAT6) downstream of IL-4 or TSG6 signaling. Adoptive transfer of Gas6-depleted BMMФs failed to clear PMNs in lungs following LPS challenge and mice showed severely defective resolution of lung injury. Thus, activation of STAT6-mediated Gas6 expression during macrophage phenotype transition resulting in efferocytosis of PMNs plays a crucial role in the resolution of inflammatory lung injury.


Assuntos
Apoptose , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Macrófagos/metabolismo , Neutrófilos/metabolismo , Fator de Transcrição STAT6/metabolismo , Transferência Adotiva , Animais , Moléculas de Adesão Celular/metabolismo , Feminino , Interleucina-4/metabolismo , Lipopolissacarídeos , Lesão Pulmonar/patologia , Masculino , Camundongos Endogâmicos C57BL , Fagocitose , Fenótipo , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia
2.
Curr Eye Res ; 44(8): 873-881, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30935217

RESUMO

Purpose: To investigate the therapeutic efficacy of tumor necrosis factor (TNF)-α stimulated gene/protein 6 (TSG-6) in a rat model of corneal alkali injury. Methods: Corneal alkali injury was produced by placing an NaOH-soaked filter paper disk on the central cornea of the right eye of an anesthetized male Lewis (LEW/Crl) rat. Recombinant human TSG-6, or an equal volume of phosphate-buffered saline (PBS), was administered intravenously (IV), by anterior chamber (AC) injection, or as a topical drop. The affected eyes were photographed daily using a dissecting microscope and documented for clinical time course analysis of corneal opacification. Corneal tissue was excised at pre-determined therapeutic endpoints, with subsequent qRT-PCR or histological analyses. Results: The continuous monitoring of corneal alkali injury progression revealed TSG-6 treatments do not show sufficient effectiveness in vivo regardless of IV injection, AC injection, or topical application. Corneal opacification and neovascularization were not diminished, and gene expression was not impacted by these treatments. However, both IV and AC administration of TSG-6 significantly suppressed pro-inflammatory cytokines compared to PBS-treated eyes. Conclusion: We conclude that the therapeutic potential of TSG-6 is insufficient in a rat corneal alkali injury model.


Assuntos
Queimaduras Químicas/tratamento farmacológico , Moléculas de Adesão Celular/uso terapêutico , Doenças da Córnea/tratamento farmacológico , Modelos Animais de Doenças , Queimaduras Oculares/induzido quimicamente , Administração Oftálmica , Animais , Câmara Anterior/efeitos dos fármacos , Queimaduras Químicas/etiologia , Queimaduras Químicas/patologia , Moléculas de Adesão Celular/administração & dosagem , Doenças da Córnea/induzido quimicamente , Doenças da Córnea/patologia , Infusões Intravenosas , Injeções Intraoculares , Masculino , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Hidróxido de Sódio
3.
Mol Ther Methods Clin Dev ; 13: 67-76, 2019 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-30719485

RESUMO

Extracellular vesicles (EVs) play key roles in cell biology and may provide new clinical diagnostics and therapies. However, it has proven difficult to develop protocols for their purification and characterization. One of the major barriers in the field has been a lack of convenient assays for their bioactivity. Developing assays has not been a trivial matter, because of the heterogeneity of EVs, the multiple activities they demonstrate, and the uncertainty about their modes of action. Therefore, it is likely that multiple assays for their activities are needed. One important assay will be for the anti-inflammatory activity observed in mice after administration of the small EVs commonly referred to as exosomes. We developed an assay for the anti-inflammatory activity of exosomes with a line of mouse macrophages. The assay makes it possible to rank different preparations of exosomes by their anti-inflammatory activity, and their ranking predicts their efficacy in suppressing LPS-stimulated inflammation in mice. The assay is convenient for comparing multiple samples and, therefore, should be useful in developing protocols for the purification and characterization of anti-inflammatory exosomes.

4.
Int J Mol Sci ; 21(1)2019 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-31888012

RESUMO

Extracellular vesicles (EVs) derived from human bone marrow mesenchymal stem cells (hMSCs) have great promise as biologics to treat neurological and neurodegenerative conditions due to their robust antiinflammatory and neuroprotective properties. Besides, intranasal (IN) administration of EVs has caught much attention because the procedure is noninvasive, amenable for repetitive dispensation, and leads to a quick penetration of EVs into multiple regions of the forebrain. Nonetheless, it is unknown whether brain injury-induced signals are essential for the entry of IN-administered EVs into different brain regions. Therefore, in this study, we investigated the distribution of IN-administered hMSC-derived EVs into neurons and microglia in the intact and status epilepticus (SE) injured rat forebrain. Ten billion EVs labeled with PKH26 were dispensed unilaterally into the left nostril of naïve rats, and rats that experienced two hours of kainate-induced SE. Six hours later, PKH26 + EVs were quantified from multiple forebrain regions using serial brain sections processed for different neural cell markers and confocal microscopy. Remarkably, EVs were seen bilaterally in virtually all regions of intact and SE-injured forebrain. The percentage of neurons incorporating EVs were comparable for most forebrain regions. However, in animals that underwent SE, a higher percentage of neurons incorporated EVs in the hippocampal CA1 subfield and the entorhinal cortex, the regions that typically display neurodegeneration after SE. In contrast, the incorporation of EVs by microglia was highly comparable in every region of the forebrain measured. Thus, unilateral IN administration of EVs is efficient for delivering EVs bilaterally into neurons and microglia in multiple regions in the intact or injured forebrain. Furthermore, incorporation of EVs by neurons is higher in areas of brain injury, implying that injury-related signals likely play a role in targeting of EVs into neurons, which may be beneficial for EV therapy in various neurodegenerative conditions including traumatic brain injury, stroke, multiple sclerosis, and Alzheimer's disease.


Assuntos
Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/citologia , Prosencéfalo/citologia , Estado Epiléptico/terapia , Administração Intranasal , Animais , Células Cultivadas , Modelos Animais de Doenças , Vesículas Extracelulares/química , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Compostos Orgânicos/farmacologia , Prosencéfalo/metabolismo , Ratos , Estado Epiléptico/metabolismo , Resultado do Tratamento
5.
Proc Natl Acad Sci U S A ; 116(1): 287-296, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30559206

RESUMO

Medial ganglionic eminence (MGE)-like interneuron precursors derived from human induced pluripotent stem cells (hiPSCs) are ideal for developing patient-specific cell therapy in temporal lobe epilepsy (TLE). However, their efficacy for alleviating spontaneous recurrent seizures (SRS) or cognitive, memory, and mood impairments has never been tested in models of TLE. Through comprehensive video- electroencephalographic recordings and a battery of behavioral tests in a rat model, we demonstrate that grafting of hiPSC-derived MGE-like interneuron precursors into the hippocampus after status epilepticus (SE) greatly restrained SRS and alleviated cognitive, memory, and mood dysfunction in the chronic phase of TLE. Graft-derived cells survived well, extensively migrated into different subfields of the hippocampus, and differentiated into distinct subclasses of inhibitory interneurons expressing various calcium-binding proteins and neuropeptides. Moreover, grafting of hiPSC-MGE cells after SE mediated several neuroprotective and antiepileptogenic effects in the host hippocampus, as evidenced by reductions in host interneuron loss, abnormal neurogenesis, and aberrant mossy fiber sprouting in the dentate gyrus (DG). Furthermore, axons from graft-derived interneurons made synapses on the dendrites of host excitatory neurons in the DG and the CA1 subfield of the hippocampus, implying an excellent graft-host synaptic integration. Remarkably, seizure-suppressing effects of grafts were significantly reduced when the activity of graft-derived interneurons was silenced by a designer drug while using donor hiPSC-MGE cells expressing designer receptors exclusively activated by designer drugs (DREADDs). These results implied the direct involvement of graft-derived interneurons in seizure control likely through enhanced inhibitory synaptic transmission. Collectively, the results support a patient-specific MGE cell grafting approach for treating TLE.


Assuntos
Encéfalo/embriologia , Epilepsia/cirurgia , Hipocampo/cirurgia , Células-Tronco Pluripotentes Induzidas/transplante , Estado Epiléptico/cirurgia , Afeto , Animais , Região CA1 Hipocampal/fisiologia , Cognição , Giro Denteado/fisiologia , Epilepsia do Lobo Temporal/cirurgia , Humanos , Masculino , Ratos , Ratos Endogâmicos F344 , Convulsões/cirurgia , Sinapses/fisiologia
6.
Mol Ther ; 26(1): 162-172, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29301108

RESUMO

The cornea is a transparent tissue devoid of blood and lymphatic vessels. However, various inflammatory conditions can cause hemangiogenesis and lymphangiogenesis in the cornea, compromising transparency and visual acuity. Mesenchymal stem/stromal cells (MSCs) have therapeutic potentials in a variety of diseases because of anti-inflammatory properties. Herein, we investigated the effects of MSCs on corneal angiogenesis using a model of suture-induced inflammatory corneal neovascularization. Data demonstrated that an intravenous administration of MSCs suppressed corneal inflammation and neovascularization, inhibiting both hemangiogenesis and lymphangiogenesis. MSCs reduced the levels of vascular endothelial growth factor (VEGF)-C, VEGF-D, Tek, MRC1, and MRC2 in the cornea, which are expressed by pro-angiogenic macrophages. Moreover, the number of CD11b+ monocytes/macrophages in the cornea, spleen, peripheral blood, and draining lymph nodes was decreased by MSCs. Depletion of circulating CD11b+ monocytes by blocking antibodies replicated the effects of MSCs. Importantly, knockdown of tumor necrosis factor alpha (TNF-α)-stimulated gene/protein 6 (TSG-6) in MSCs abrogated the effects of MSCs in inhibiting corneal hemangiogenesis and lymphangiogenesis and monocyte/macrophage infiltration. Together, the results suggest that MSCs inhibit inflammatory neovascularization in the cornea by suppressing pro-angiogenic monocyte/macrophage recruitment in a TSG-6-dependent manner.


Assuntos
Moléculas de Adesão Celular/metabolismo , Córnea/metabolismo , Ceratite/imunologia , Ceratite/metabolismo , Linfangiogênese , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Biomarcadores , Biópsia , Linhagem Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Ceratite/patologia , Linfonodos , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Transcrição Gênica
7.
Mol Ther ; 25(8): 1748-1756, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28647464

RESUMO

Much of what we know about immunology suggests that little is to be gained from experiments in which human cells are administered to immunocompetent mice. Multiple reports have demonstrated that this common assumption does not hold for experiments with human mesenchymal stem/stromal cells (hMSCs). The data demonstrate that hMSCs can suppress immune responses to a variety of stimuli in immunocompetent mice by a range of different mechanisms that are similar to those employed by mouse MSCs. Therefore, further experiments with hMSCs in mice will make it possible to generate preclinical data that will improve both the efficacy and safety of the clinical trials with the cells that are now in progress.


Assuntos
Tolerância Imunológica , Imunomodulação , Células-Tronco Mesenquimais/metabolismo , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Imunidade , Células-Tronco Mesenquimais/citologia , Camundongos , Especificidade da Espécie
8.
Stem Cell Reports ; 8(5): 1214-1225, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28494937

RESUMO

Accumulating evidence shows that extracellular vesicles (EVs) produced by mesenchymal stem/stromal cells (MSCs) exert their therapeutic effects in several disease models. We previously demonstrated that MSCs suppress autoimmunity in models of type 1 diabetes (T1D) and experimental autoimmune uveoretinitis (EAU). Therefore, here, we investigated the therapeutic potential of MSC-derived EVs using our established mouse models for autoimmune diseases affecting the pancreas and the eye: T1D and EAU. The data demonstrate that MSC-derived EVs effectively prevent the onset of disease in both T1D and EAU. In addition, the mixed lymphocyte reaction assay with MSC-derived EVs indicated that EVs inhibit activation of antigen-presenting cells and suppress development of T helper 1 (Th1) and Th17 cells. These results raise the possibility that MSC-derived EVs may be an alternative to cell therapy for autoimmune disease prevention.


Assuntos
Autoimunidade , Diabetes Mellitus Tipo 1/terapia , Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/citologia , Retinite/terapia , Uveíte/terapia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Vesículas Extracelulares/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos SCID , Retinite/imunologia , Células Th1/imunologia , Células Th17/imunologia , Uveíte/imunologia
9.
Proc Natl Acad Sci U S A ; 114(17): E3536-E3545, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28396435

RESUMO

Status epilepticus (SE), a medical emergency that is typically terminated through antiepileptic drug treatment, leads to hippocampus dysfunction typified by neurodegeneration, inflammation, altered neurogenesis, as well as cognitive and memory deficits. Here, we examined the effects of intranasal (IN) administration of extracellular vesicles (EVs) secreted from human bone marrow-derived mesenchymal stem cells (MSCs) on SE-induced adverse changes. The EVs used in this study are referred to as A1-exosomes because of their robust antiinflammatory properties. We subjected young mice to pilocarpine-induced SE for 2 h and then administered A1-exosomes or vehicle IN twice over 24 h. The A1-exosomes reached the hippocampus within 6 h of administration, and animals receiving them exhibited diminished loss of glutamatergic and GABAergic neurons and greatly reduced inflammation in the hippocampus. Moreover, the neuroprotective and antiinflammatory effects of A1-exosomes were coupled with long-term preservation of normal hippocampal neurogenesis and cognitive and memory function, in contrast to waned and abnormal neurogenesis, persistent inflammation, and functional deficits in animals receiving vehicle. These results provide evidence that IN administration of A1-exosomes is efficient for minimizing the adverse effects of SE in the hippocampus and preventing SE-induced cognitive and memory impairments.


Assuntos
Exossomos/transplante , Transtornos da Memória/terapia , Células-Tronco Mesenquimais/metabolismo , Neurogênese , Estado Epiléptico/terapia , Administração Intranasal , Animais , Linhagem Celular , Exossomos/metabolismo , Exossomos/patologia , Humanos , Masculino , Transtornos da Memória/metabolismo , Transtornos da Memória/patologia , Transtornos da Memória/fisiopatologia , Células-Tronco Mesenquimais/patologia , Camundongos , Estado Epiléptico/metabolismo , Estado Epiléptico/patologia , Estado Epiléptico/fisiopatologia
10.
Cytotherapy ; 19(1): 28-35, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27840134

RESUMO

BACKGROUND AIMS: Mesenchymal stromal cells (MSCs) offer tremendous potential for therapeutic applications for inflammatory diseases. However, tissue-derived MSCs, such as bone marrow-derived MSCs (BM-MSCs), have considerable donor variations and limited expandability. It was recently demonstrated that MSCs derived from induced pluripotent stem cells (iPSC-MSCs) have less pro-tumor potential and greater expandability of homogenous cell population. In this study, we investigated the anti-inflammatory effects and mechanism of iPSC-MSCs in a murine model of chemical and mechanical injury to the cornea and compared the effects with those of BM-MSCs. METHODS: To create an injury, ethanol was applied to the corneal surface in mice, and the corneal epithelium was removed with a blade. Immediately after injury, mice received an intravenous injection of (i) iPSC-MSCs, (ii) BM-MSCs or (iii) vehicle. Clinical, histological and molecular assays were performed in the cornea to evaluate inflammation. RESULTS: We found that corneal opacity was significantly reduced by iPSC-MSCs or BM-MSCs. Histological examination revealed that the swelling and inflammatory infiltration in the cornea were markedly decreased in mice treated with iPSC-MSCs or BM-MSCs. Corneal levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6 were lower in iPSC-MSC- and BM-MSC-treated mice, compared with vehicle-treated controls. In contrast, iPSC-MSCs with a knockdown of the TNF-α stimulating gene (TSG)-6 did not suppress the levels of inflammatory cytokines and failed to reduce corneal opacity. CONCLUSIONS: Together these data demonstrate that iPSC-MSCs exert therapeutic effects in the cornea by reducing inflammation in part through the expression of TSG-6, and the effects are similar to those seen with BM-MSCs.


Assuntos
Lesões da Córnea/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Mesenquimais/citologia , Animais , Células da Medula Óssea/citologia , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Lesões da Córnea/metabolismo , Opacidade da Córnea/patologia , Opacidade da Córnea/terapia , Modelos Animais de Doenças , Células-Tronco Pluripotentes Induzidas/transplante , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Ceratite/patologia , Ceratite/terapia , Transplante de Células-Tronco Mesenquimais , Camundongos Endogâmicos BALB C , Fator de Necrose Tumoral alfa/metabolismo
11.
Cytotherapy ; 19(1): 1-8, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27769637

RESUMO

From the outset, it was apparent that developing new therapies with mesenchymal stem/stromal cells (MSCs) was not a simple or easy task. Among the earliest experiments was administration of MSCs from normal mice to transgenic mice that developed brittle bones because they expressed a mutated gene for type 1 collagen isolated from a patient with osteogenesis imperfecta. The results prompted a clinical trial of MSCs in patients with severe osteogenesis imperfecta. Subsequent work by large numbers of scientists and clinicians has established that, with minor exceptions, MSCs do not engraft or differentiate to a large extent in vivo. Instead the cells produce beneficial effects in a large number of animal models and some clinical trials by secreting paracrine factors and extracellular vesicles in a "hit and run" scenario. The field faces a number of challenges, but the results indicate that we are on the way to effective therapies for millions of patients who suffer from devastating diseases.


Assuntos
Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Colágeno Tipo I/genética , Humanos , Células-Tronco Mesenquimais/fisiologia , Camundongos Transgênicos , Osteogênese Imperfeita/genética
12.
Proc Natl Acad Sci U S A ; 113(50): E8151-E8158, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911817

RESUMO

TNFα-stimulated gene-6 (TSG6), a 30-kDa protein generated by activated macrophages, modulates inflammation; however, its mechanism of action and role in the activation of macrophages are not fully understood. Here we observed markedly augmented LPS-induced inflammatory lung injury and mortality in TSG6-/- mice compared with WT (TSG6+/+) mice. Treatment of mice with intratracheal instillation of TSG6 prevented LPS-induced lung injury and neutrophil sequestration, and increased survival in mice. We found that TSG6 inhibited the association of TLR4 with MyD88, thereby suppressing NF-κB activation. TSG6 also prevented the expression of proinflammatory proteins (iNOS, IL-6, TNFα, IL-1ß, and CXCL1) while increasing the expression of anti-inflammatory proteins (CD206, Chi3l3, IL-4, and IL-10) in macrophages. This shift was associated with suppressed activation of proinflammatory transcription factors STAT1 and STAT3. In addition, we observed that LPS itself up-regulated the expression of TSG6 in TSG6+/+ mice, suggesting an autocrine role for TSG6 in transitioning macrophages. Thus, TSG6 functions by converting macrophages from a proinflammatory to an anti-inflammatory phenotype secondary to suppression of TLR4/NF-κB signaling and STAT1 and STAT3 activation.


Assuntos
Moléculas de Adesão Celular/imunologia , Lesão Pulmonar/prevenção & controle , Macrófagos/imunologia , Animais , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/genética , Reprogramação Celular/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/imunologia , Lipopolissacarídeos/toxicidade , Pulmão/irrigação sanguínea , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Ativação de Macrófagos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Fenótipo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
13.
Proc Natl Acad Sci U S A ; 113(42): E6447-E6456, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27698134

RESUMO

Patients with breast cancer often develop malignant regrowth of residual drug-resistant dormant tumor cells years after primary treatment, a process defined as cancer relapse. Deciphering the causal basis of tumor dormancy therefore has obvious therapeutic significance. Because cancer cell behavior is strongly influenced by stromal cells, particularly the mesenchymal stem/stromal cells (MSCs) that are actively recruited into tumor-associated stroma, we assessed the impact of MSCs on breast cancer cell (BCC) dormancy. Using 3D cocultures to mimic the cellular interactions of an emerging tumor niche, we observed that MSCs sequentially surrounded the BCCs, promoted formation of cancer spheroids, and then were internalized/degraded through a process resembling the well-documented yet ill-defined clinical phenomenon of cancer cell cannibalism. This suspected feeding behavior was less appreciable in the presence of a rho kinase inhibitor and in 2D monolayer cocultures. Notably, cannibalism of MSCs enhanced survival of BCCs deprived of nutrients but suppressed their tumorigenicity, together suggesting the cancer cells entered dormancy. Transcriptome profiles revealed that the resulting BCCs acquired a unique molecular signature enriched in prosurvival factors and tumor suppressors, as well as inflammatory mediators that demarcate the secretome of senescent cells, also referred to as the senescence-associated secretory phenotype. Overall, our results provide intriguing evidence that cancer cells under duress enter dormancy after cannibalizing MSCs. Importantly, our practical 3D coculture model could provide a valuable tool to understand the antitumor activity of MSCs and cell cannibalism further, and therefore open new therapeutic avenues for the prevention of cancer recurrence.


Assuntos
Citofagocitose , Células-Tronco Mesenquimais/metabolismo , Neoplasias/metabolismo , Fase de Repouso do Ciclo Celular , Animais , Biomarcadores , Comunicação Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Citocinas , Modelos Animais de Doenças , Feminino , Expressão Gênica , Genes Reporter , Xenoenxertos , Humanos , Camundongos , Neoplasias/etiologia , Neoplasias/patologia , Fenótipo , Transdução de Sinais , Esferoides Celulares , Estresse Fisiológico , Células Tumorais Cultivadas
14.
Transl Res ; 177: 127-142, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27469269

RESUMO

Stanniocalcin-1 (STC-1) is a multifunctional glycoprotein with antioxidant and anti-inflammatory properties. Ischemic myocardial necrosis generates "danger" signals that perpetuate detrimental inflammatory reactions often involving monocyte recruitment and their subsequent differentiation into proinflammatory macrophages. Therefore, we evaluated the effects of recombinant STC-1 (rSTC-1) on monocyte phenotype and in a mouse model of myocardial infarction. Using an established protocol to differentiate human monocytes into macrophages, we demonstrated that rSTC-1 did not alter morphology of the differentiated cells, toll-like receptor (TLR) 4 expression, or expression of the myeloid cell marker CD11b. However, rSTC-1 treatment before differentiation attenuated the rise in the expression of CD14, a TLR4 coreceptor and pathogen sensor that propagates innate immune responses, and suppressed levels of inflammatory cytokines produced by the differentiated cells in response to the CD14-TLR4 ligand lipopolysaccharide. Moreover, rSTC-1 treatment reduced CD14 expression in monocytes stimulated with endogenous danger signals. Interestingly, the effects of rSTC-1 on CD14 expression were not reproduced by a superoxide dismutase mimetic. In mice with induced myocardial infarcts, intravenous administration of rSTC-1 decreased CD14 expression in the heart as well as levels of tumor necrosis factor alpha, C-X-C motif ligand 2, interleukin 1 beta, and myeloperoxidase. It also suppressed the formation of scar tissue while enhancing cardiac function. The data suggests that one of the beneficial effects of STC-1 might be attributed to suppression of CD14 on recruited monocytes and macrophages that limits their inflammatory response. STC-1 may be a promising therapy to protect the heart and other tissues from ischemic injury.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glicoproteínas/farmacologia , Glicoproteínas/uso terapêutico , Inflamação/patologia , Macrófagos/patologia , Monócitos/patologia , Isquemia Miocárdica/tratamento farmacológico , Animais , Biomarcadores/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cicatriz/patologia , Citocinas/metabolismo , Testes de Função Cardíaca/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Isquemia Miocárdica/diagnóstico por imagem , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Células U937
15.
Stem Cell Res Ther ; 7: 27, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26864573

RESUMO

BACKGROUND: Mesenchymal stem/progenitor cells (MSC) have shown beneficial effects in many models of disease in part by modulating excessive inflammatory and immune responses. Frequently the beneficial effects of MSC persist long after their disappearance from host tissues, suggesting that MSC interact with intermediate cells in the host that relay or amplify their effects. The cells have usually been injected intravenously, but beneficial effects have also been reported with intraperitoneal (IP) injection of MSC. However the fate of IP injection of MSC has not been examined. METHODS: The fate of the human MSC injected IP into immune-competent mice was studied. In vivo imaging was used to track green fluorescent protein-labeled MSC in the peritoneal cavity. In addition, their retention in peritoneal tissues was measured by real-time polymerase chain reaction for human GAPDH mRNA. To describe the effects of human MSC on the immune system of the peritoneum, the peritoneal lavage, omentum, lymph nodes and mesenteric tissues were collected. Flow cytometry was used to evaluate the immune cell populations, while cytokine/chemokine production was measured by real-time polymerase chain reaction and enzyme-linked immunosorbent assay. Challenge with lipopolysaccharide at 3 days after the administration of MSC was used to evaluate the preconditioning of the immune system. RESULTS: Within 20 min, single MSC were no longer detected in peritoneal lavage fluid. Instead they were recovered as aggregates of varying size that contained mouse macrophages and a few B220+ lymphocytes. After 1 day, most of the aggregates containing live MSC were attached to sites throughout the peritoneal cavity including the omentum and mesentery. Less than 0.05 % of the live injected cells were detected in the spleen and jejunal lymph nodes. In all locations, MSC colocalized with mouse macrophages and B220+ lymphocytes. Attachment to the omentum and mesentery was accompanied by the recruitment of immune cells and changes in the production of a series of mouse cytokines. A similar increase in mouse cytokines in the peritoneum was seen after IP injections of human fibroblasts. CONCLUSIONS: IP injected human MSC rapidly formed aggregates with mouse macrophages and B220+ lymphocytes and attached to the walls of the peritoneal cavity. The formation of the aggregates probably limits access of the cells to the systemic circulation.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Animais , Adesão Celular , Agregação Celular , Citocinas/biossíntese , Citocinas/metabolismo , Humanos , Infusões Parenterais , Antígenos Comuns de Leucócito/metabolismo , Lipopolissacarídeos/farmacologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peritônio/citologia , Peritônio/imunologia
16.
PLoS One ; 11(1): e0147553, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26793973

RESUMO

TNF-α stimulated gene/protein 6 (TNFAIP6/TSG-6) is a multifunctional protein that has a number of potential therapeutic applications. Experiments and clinical trials with TSG-6, however, have been limited by the technical difficulties of producing the recombinant protein. We prepared stable clones of CHO cells that expressed recombinant human TSG-6 (rhTSG-6) as a secreted glycoprotein. Paradoxically, both cell number and protein production decreased dramatically when the clones were expanded. The decreases occurred because the protein aggregated the synthesizing CHO cells by binding to the brush border of hyaluronan that is found around many cultured cells. In addition, the rhTSG-6 readily self-aggregated. To address these problems, we added to the medium an inhibitor of hyaluronan synthesis and heparin to compete with the binding of TSG-6 to hyaluronan. Also, we optimized the composition of the culture medium, and transferred the CHO cells from a spinner culture system to a bioreactor that controlled pH and thereby decreased pH-dependent binding properties of the protein. With these and other improvements in the culture conditions, we obtained 57.0 mg ± 9.16 S.D. of rhTSG-6 in 5 or 6 liter of medium. The rhTSG-6 accounted for 18.0% ± 3.76 S.D. of the total protein in the medium. We then purified the protein with a Ni-chelate column that bound the His tag engineered into the C-terminus of the protein followed by an anion exchange column. The yield of the purified monomeric rhTSG-6 was 4.1 mg to 5.6 mg per liter of culture medium. After intravenous injection into mice, the protein had a longer plasma half-life than commercially available rhTSG-6 isolated from a mammalian cell lysate, apparently because it was recovered as a secreted glycoprotein. The bioactivity of the rhTSG-6 in suppressing inflammation was demonstrated in a murine model.


Assuntos
Células-Tronco Adultas/citologia , Moléculas de Adesão Celular/química , Inflamação/prevenção & controle , Proteínas Recombinantes/química , Células-Tronco Adultas/metabolismo , Animais , Moléculas de Adesão Celular/administração & dosagem , Moléculas de Adesão Celular/metabolismo , Cricetinae , Cricetulus , Meia-Vida , Humanos , Ácido Hialurônico/metabolismo , Inflamação/induzido quimicamente , Inflamação/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo
17.
Matrix Biol ; 51: 7-13, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26807758

RESUMO

Fibrosis and scarring are the end stage of many disease processes. In effect, the collagen fibers that initially provide a necessary strength during the repair of injured tissues are frequently synthesized in excessive amounts and become irreversible fibrotic deposits that limit regeneration of the endogenous cells of a tissue. This review will focus on the potential of mesenchymal stem/stromal cells for treatment of fibrotic diseases, with emphasis on the role of TSG-6 as a mediator of anti-inflammatory effects.


Assuntos
Moléculas de Adesão Celular/genética , Fibrose/genética , Inflamação/genética , Transplante de Células-Tronco Mesenquimais , Moléculas de Adesão Celular/uso terapêutico , Terapia Baseada em Transplante de Células e Tecidos/métodos , Cicatriz/tratamento farmacológico , Cicatriz/patologia , Colágeno/genética , Fibrose/patologia , Fibrose/terapia , Humanos , Inflamação/patologia , Inflamação/terapia , Regeneração/genética
18.
Proc Natl Acad Sci U S A ; 113(1): 158-63, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26699483

RESUMO

Intravenously administered mesenchymal stem/stromal cells (MSCs) engraft only transiently in recipients, but confer long-term therapeutic benefits in patients with immune disorders. This suggests that MSCs induce immune tolerance by long-lasting effects on the recipient immune regulatory system. Here, we demonstrate that i.v. infusion of MSCs preconditioned lung monocytes/macrophages toward an immune regulatory phenotype in a TNF-α-stimulated gene/protein (TSG)-6-dependent manner. As a result, mice were protected against subsequent immune challenge in two models of allo- and autoimmune ocular inflammation: corneal allotransplantation and experimental autoimmune uveitis (EAU). The monocytes/macrophages primed by MSCs expressed high levels of MHC class II, B220, CD11b, and IL-10, and exhibited T-cell-suppressive activities independently of FoxP3(+) regulatory T cells. Adoptive transfer of MSC-induced B220(+)CD11b(+) monocytes/macrophages prevented corneal allograft rejection and EAU. Deletion of monocytes/macrophages abrogated the MSC-induced tolerance. However, MSCs with TSG-6 knockdown did not induce MHC II(+)B220(+)CD11b(+) cells, and failed to attenuate EAU. Therefore, the results demonstrate a mechanism of the MSC-mediated immune modulation through induction of innate immune tolerance that involves monocytes/macrophages.


Assuntos
Autoimunidade/imunologia , Tolerância Imunológica/imunologia , Pulmão/imunologia , Células-Tronco Mesenquimais/imunologia , Monócitos/imunologia , Uveíte/imunologia , Administração Intravenosa , Animais , Antígeno CD11b/imunologia , Moléculas de Adesão Celular/genética , Córnea/imunologia , Transplante de Córnea , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Genes MHC da Classe II/imunologia , Sobrevivência de Enxerto/imunologia , Tolerância Imunológica/genética , Interleucina-10/imunologia , Antígenos Comuns de Leucócito/imunologia , Macrófagos/imunologia , Transplante de Células-Tronco Mesenquimais , Camundongos , Linfócitos T Reguladores/imunologia
19.
Proc Natl Acad Sci U S A ; 113(1): 170-5, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26699510

RESUMO

Extracellular vesicles (EVs) secreted by cells present an attractive strategy for developing new therapies, but progress in the field is limited by several issues: The quality of the EVs varies with the type and physiological status of the producer cells; protocols used to isolate the EVs are difficult to scale up; and assays for efficacy are difficult to develop. In the present report, we have addressed these issues by using human mesenchymal stem/stromal cells (MSCs) that produce EVs when incubated in a protein-free medium, preselecting the preparations of MSCs with a biomarker for their potency in modulating inflammation, incubating the cells in a chemically defined protein-free medium that provided a stable environment, isolating the EVs with a scalable chromatographic procedure, and developing an in vivo assay for efficacy of the cells in suppressing neuroinflammation after traumatic brain injury (TBI) in mice. In addition, we demonstrate that i.v. infusion of the isolated EVs shortly after induction of TBI rescued pattern separation and spatial learning impairments 1 mo later.


Assuntos
Lesões Encefálicas/complicações , Transtornos Cognitivos/terapia , Encefalite/terapia , Vesículas Extracelulares/química , Células-Tronco Mesenquimais/química , Animais , Biomarcadores/análise , Lesões Encefálicas/psicologia , Células Cultivadas , Cromatografia por Troca Iônica , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/psicologia , Meios de Cultura Livres de Soro , Encefalite/etiologia , Encefalite/psicologia , Humanos , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Aprendizagem Espacial , Tetraspanina 28/análise , Tetraspanina 30/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA