Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 720
Filtrar
1.
Gene Ther ; 30(3-4): 309-322, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35931871

RESUMO

A primary goal in transplantation medicine is the induction of a tolerogenic environment for prevention of transplant rejection without the need for long-term pharmacological immunosuppression. Generation of alloantigen-specific regulatory T cells (Tregs) by transduction with chimeric antigen receptors (CARs) is a promising strategy to achieve this goal. This publication reports the preclinical characterization of Tregs (TR101) transduced with a human leukocyte antigen (HLA)-A*02 CAR lentiviral vector (TX200) designated to induce immunosuppression of allograft-specific effector T cells in HLA-A*02-negative recipients of HLA-A*02-positive transplants. In vitro results demonstrated specificity, immunosuppressive function, and safety of TX200-TR101. In NOD scid gamma (NSG) mice, TX200-TR101 prevented graft-versus-host disease (GvHD) in a xenogeneic GvHD model and TX200-TR101 Tregs localized to human HLA-A*02-positive skin transplants in a transplant model. TX200-TR101 persisted over the entire duration of a 3-month study in humanized HLA-A*02 NSG mice and remained stable, without switching to a proinflammatory phenotype. Concomitant tacrolimus did not impair TX200-TR101 Treg survival or their ability to inhibit peripheral blood mononuclear cell (PBMC) engraftment. These data demonstrate that TX200-TR101 is specific, stable, efficacious, and safe in preclinical models, and provide the basis for a first-in-human study.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Órgãos , Receptores de Antígenos Quiméricos , Camundongos , Animais , Humanos , Linfócitos T Reguladores , Leucócitos Mononucleares/transplante , Antígenos HLA-A
2.
J Virol ; 96(3): e0139421, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34818071

RESUMO

Humanized mouse models are based on the engraftment of human cells in immunodeficient mouse strains, most notably the NSG strain. Most used models have a major limitation in common, the development of graft-versus-host disease (GVHD). GVHD not only introduces variabilities into the research data but also leads to animal welfare concerns. A new mouse strain, B6.129S-Rag2tm1Fwa CD47tm1Fpl Il2rgtm1Wjl/J, which lacks Rag1, IL2rg, and CD47 (triple knockout [TKO]), is resistant to GVHD development. We transplanted TKO mice with human peripheral blood mononuclear cells (PBMCs) to establish a new humanized PBMC (hu-PBMC) mouse model. A cohort of these mice was infected with HIV-1 and monitored for plasma HIV viremia and CD4+ T cell depletion. The onset and progression of GVHD were monitored by clinical signs. This study demonstrates that TKO mice transplanted with human PBMCs support engraftment of human immune cells in primary and secondary lymphoid tissues, rectum, and brain. Moreover, the TKO hu-PBMC model supports HIV-1 infection via the intraperitoneal, rectal, or vaginal route, as confirmed by robust plasma HIV viremia and CD4+ T cell depletion. Lastly, TKO mice showed a delayed onset of GVHD clinical signs (∼24 days) and exhibited significant decreases in plasma levels of tumor necrosis factor beta (TNF-ß). Based on these results, the TKO hu-PBMC mouse model not only supports humanization and HIV-1 infection but also has a delayed onset of GVHD development, making this model a valuable tool in HIV research. IMPORTANCE Currently, there is no cure or vaccine for HIV infection; thus, continued research is needed to end the HIV pandemic. While many animal models are used in HIV research, none is used more than the humanized mouse model. A major limitation with current humanized mouse models is the development of graft-versus-host disease (GVHD). Here, we describe a novel humanized-PBMC mouse model that has a delayed onset GVHD development and supports and models HIV infection comparably to well-established humanized mouse models.


Assuntos
Transplante de Células , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/etiologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Leucócitos Mononucleares/transplante , Animais , Transplante de Células/efeitos adversos , Transplante de Células/métodos , Suscetibilidade a Doenças , Feminino , HIV-1/imunologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Pesquisa , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fatores de Tempo
3.
Fertil Steril ; 116(6): 1449-1454, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34836580

RESUMO

Despite the challenges in studying recurrent implantation failure, progress is currently being made in therapeutic options to help those who suffer from recurrent implantation failure. Three of the most promising therapeutic options for recurrent implantation failure include immune therapies such as peripheral blood mononuclear cells, platelet rich plasma and subcutaneous granulocyte-colony stimulating factor.


Assuntos
Implantação do Embrião/fisiologia , Transferência Embrionária/métodos , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Imunoterapia/métodos , Plasma Rico em Plaquetas/fisiologia , Falha de Tratamento , Transferência Embrionária/tendências , Feminino , Fertilização In Vitro/métodos , Fertilização In Vitro/tendências , Humanos , Imunoterapia/tendências , Leucócitos Mononucleares/fisiologia , Leucócitos Mononucleares/transplante , Gravidez , Recidiva , Resultado do Tratamento
4.
Exp Neurol ; 346: 113864, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34520725

RESUMO

BACKGROUND AND PURPOSE: Post-stroke cognitive impairment (PSCI) has become a major public health issue, as a leading cause of dementia. The inflammation that develops soon after cerebral artery occlusion and may persist for weeks or months after stroke is a key component of PSCI. Our aim was to take advantage of the immunomodulatory properties of peripheral blood mononuclear cells (PB-MNC) stimulated with ephrin-B2/fc (PB-MNC+) for preventing PSCI. METHODS: Cortical infarct was induced by thermocoagulation of the middle cerebral artery in male diabetic mice (streptozotocin IP). PB-MNC were isolated from diabetic human donors, washed with recombinant ephrin-B2/Fc and injected into the mice intravenously on the following day. Infarct volume, sensorimotor deficit, cell death and immune cell densities were assessed on day 3. Six weeks later, cognitive assessment was performed using the Barnes maze. RESULTS: PB-MNC+ transplanted in post-stroke diabetic mice reduced the neurological deficit, infarct volume and apoptosis at D3, without modification of microglial cells, astrocytes and T-lymphocytes densities in the brain. Barnes maze assessment of memory showed that the learning, retention and reversal phases were not significantly modified by cell therapy. CONCLUSIONS: Intravenous PB-MNC+ administration the day after stroke induction in diabetic mice improved sensorimotor deficit and reduced infarct volume at the short term, but was unable to prevent long-term memory loss. To what extent diabetes impacts on cell therapy efficacy will have to be specifically investigated in the future. Including vascular risk factors systematically in preclinical studies of cell therapy will provide a comprehensive understanding of the mechanisms potentially limiting cell efficacy and also to identify good and bad responders, particularly in the long term.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Diabetes Mellitus Experimental/terapia , Efrina-B2/administração & dosagem , Leucócitos Mononucleares/transplante , Transtornos da Memória/terapia , Acidente Vascular Cerebral/terapia , Administração Intravenosa , Animais , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Experimental/psicologia , Masculino , Transtornos da Memória/fisiopatologia , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/psicologia
5.
Front Immunol ; 12: 677970, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248959

RESUMO

Objective: The contribution of sustained autologous autoantibody production by B cells to the pathogenesis of systemic sclerosis (SSc) and granulomatosis with polyangiitis (GPA) is not fully understood. To investigate this, a humanized mouse model was generated by transferring patient-derived peripheral blood mononuclear cells (PBMC) into immunocompromised mice. Methods: PBMC derived from patients with SSc and GPA as well as healthy controls (HD) were isolated, characterized by flow cytometry, and infused into Rag2-/-/IL2rg-/- mice. In addition, PBMC from SSc patients treated with rituximab were transferred into mice. Twelve weeks later, human autoantibodies were determined in blood of the recipient mice and affected tissues were analyzed for pathological changes by histology and immunohistochemistry. Results: Mice engrafted with PBMC derived from SSc patients developed autoantibodies such as antinuclear antibodies (ANA) mimicking the pattern of the respective donors. Moreover, cellular infiltrates dominated by B cells were observed in lung, kidney and muscles of the recipient mice. By contrast, PBMC derived from HD or GPA patients survived in recipient mice after transfer, but neither human autoantibodies nor inflammatory infiltrates in tissues were detected. Furthermore, these pathological changes were absent in mice transferred with PBMC from rituximab-treated SSc patients. Conclusion: This humanized mouse model is indicative for cross-reactivity of human lymphocytes to murine autoantigens and argues for a pivotal role of B cells as well as of sustained autoimmunity in the pathogenesis of SSc. It provides a powerful tool to study interstitial lung disease and so far, under-recognized disease manifestations such as myositis and interstitial nephritis.


Assuntos
Anticorpos Antinucleares/imunologia , Proteínas de Ligação a DNA/metabolismo , Granulomatose com Poliangiite/sangue , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Escleroderma Sistêmico/sangue , Adulto , Idoso , Animais , Anticorpos Antinucleares/sangue , Linfócitos B/imunologia , Reações Cruzadas , Proteínas de Ligação a DNA/genética , Feminino , Granulomatose com Poliangiite/imunologia , Humanos , Hospedeiro Imunocomprometido , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Fatores Imunológicos/uso terapêutico , Inflamação/imunologia , Subunidade gama Comum de Receptores de Interleucina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Modelos Animais , Rituximab/uso terapêutico , Escleroderma Sistêmico/tratamento farmacológico , Escleroderma Sistêmico/imunologia , Resultado do Tratamento , Adulto Jovem
6.
Front Immunol ; 12: 628059, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34122401

RESUMO

Artificial antigen-presenting cells (aAPCs) are synthetic versions of naturally occurring antigen-presenting cells (APCs) that, similar to natural APCs, promote efficient T effector cell responses in vitro. This report describes a method to produce acellular tolerogenic aAPCs made of biodegradable poly lactic-co-glycolic acid (PLGA) nanoparticles (NPs) and encapsulating IL-2 and TGF-ß for a paracrine release to T cells. We document that these aAPCs can induce both human CD4+ and CD8+ T cells to become FoxP3+ T regulatory cells (Tregs). The aAPC NP-expanded human Tregs are functional in vitro and can modulate systemic autoimmunity in vivo in humanized NSG mice. These findings establish a proof-of-concept to use PLGA NPs as aAPCs for the induction of human Tregs in vitro and in vivo, highlighting the immunotherapeutic potential of this targeted approach to repair IL-2 and/or TGF-ß defects documented in certain autoimmune diseases such as systemic lupus erythematosus.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Doença Enxerto-Hospedeiro/prevenção & controle , Interleucina-2/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/transplante , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Linfócitos T Reguladores/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/metabolismo , Humanos , Interleucina-2/química , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Estudo de Prova de Conceito , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/química
7.
Front Immunol ; 12: 687715, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34177940

RESUMO

The outcome of organ transplantation is largely dictated by selection of a well-matched donor, which results in less chance of graft rejection. An allogeneic immune response is the main immunological barrier for successful organ transplantation. Donor and recipient human leukocyte antigen (HLA) mismatching diminishes outcomes after solid organ transplantation. The current evaluation of HLA incompatibility does not provide information on the immunogenicity of individual HLA mismatches and impact of non-HLA-related alloantigens, especially in vivo. Here we demonstrate a new method for analysis of alloimmune responsiveness between donor and recipient in vivo by introducing a humanized mouse model. Using molecular, cellular, and genomic analyses, we demonstrated that a recipient's personalized humanized mouse provided the most sensitive assessment of allogeneic responsiveness to potential donors. In our study, HLA typing provided a better recipient-donor match for one donor among two related donors. In contrast, assessment of an allogeneic response by mixed lymphocyte reaction (MLR) was indistinguishable between these donors. We determined that, in the recipient's humanized mouse model, the donor selected by HLA typing induced the strongest allogeneic response with markedly increased allograft rejection markers, including activated cytotoxic Granzyme B-expressing CD8+ T cells. Moreover, the same donor induced stronger upregulation of genes involved in the allograft rejection pathway as determined by transcriptome analysis of isolated human CD45+cells. Thus, the humanized mouse model determined the lowest degree of recipient-donor alloimmune response, allowing for better selection of donor and minimized immunological risk of allograft rejection in organ transplantation. In addition, this approach could be used to evaluate the level of alloresponse in allogeneic cell-based therapies that include cell products derived from pluripotent embryonic stem cells or adult stem cells, both undifferentiated and differentiated, all of which will produce allogeneic immune responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/prevenção & controle , Antígenos HLA/imunologia , Teste de Histocompatibilidade , Histocompatibilidade , Leucócitos Mononucleares/transplante , Transplante de Órgãos , Baço/imunologia , Tolerância ao Transplante , Animais , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Bases de Dados Genéticas , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Sobrevivência de Enxerto , Antígenos HLA/genética , Humanos , Isoanticorpos/metabolismo , Leucócitos Mononucleares/imunologia , Teste de Cultura Mista de Linfócitos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante de Órgãos/efeitos adversos , Fenótipo , Valor Preditivo dos Testes , Baço/metabolismo , Transcriptoma , Transplante Homólogo
8.
Signal Transduct Target Ther ; 6(1): 174, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33958574

RESUMO

Severely immunosuppressed AIDS patients with recurrent opportunistic infections (OIs) represent an unmet medical need even in the era of antiretroviral therapy (ART). Here we report the development of a human leukocyte antigen (HLA)-mismatched allogeneic adaptive immune therapy (AAIT) for severely immunosuppressed AIDS patients. Twelve severely immunosuppressed AIDS patients with severe OIs were enrolled in this single-arm study. Qualified donors received subcutaneous recombinant granulocyte-colony-stimulating factor twice daily for 4-5 days to stimulate hematopoiesis. Peripheral blood mononuclear cells were collected from these donors via leukapheresis and transfused into the coupled patients. Clinical, immunological, and virological parameters were monitored during a 12-month follow-up period. We found AAIT combined with ART was safe and well-tolerated at the examined doses and transfusion regimen in all 12 patients. Improvements in clinical symptoms were evident throughout the study period. All patients exhibited a steady increase of peripheral CD4+ T cells from a median 10.5 to 207.5 cells/µl. Rapid increase in peripheral CD8+ T-cell count from a median 416.5 to 1206.5 cells/µl was found in the first 90 days since initiation of AAIT. In addition, their inflammatory cytokine levels and HIV RNA viral load decreased. A short-term microchimerism with donor cells was found. There were no adverse events associated with graft-versus-host disease throughout the study period. Overall, AAIT treatment was safe, and might help severely immunosuppressed AIDS patients to achieve a better immune restoration. A further clinical trial with control is necessary to confirm the efficacy of AAIT medication.


Assuntos
Síndrome de Imunodeficiência Adquirida , Transferência Adotiva , HIV-1/imunologia , Antígenos HLA/imunologia , Leucócitos Mononucleares , Síndrome de Imunodeficiência Adquirida/imunologia , Síndrome de Imunodeficiência Adquirida/patologia , Síndrome de Imunodeficiência Adquirida/terapia , Adolescente , Adulto , Aloenxertos , Feminino , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/transplante , Masculino , Pessoa de Meia-Idade
9.
J Reprod Immunol ; 145: 103323, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33878637

RESUMO

The purpose of this study was to assess whether intrauterine administration of peripheral blood mononuclear cells (PBMCs) activated by human chorionic gonadotropin (hCG) could improve the pregnancy and live birth rates in women with repeated implantation failure (RIF), and whether the parameters of co-culture of hCG and PBMCs would affect the clinical outcomes. Six databases (PubMed, Ovid, Medline, NCBI, Cqvip and Wanfang) were searched up to October 2020 by two independent reviewers. Seven studies were included according to specific inclusion and exclusion criteria. A meta-analysis showed that the pregnancy and live birth rates were significantly increased in the case group compared with the control group (odds ratio [OR]: 3.43, 95 % confidence interval [CI]: 1.78-6.61; P = 0.0002 and OR: 2.79, 95 % CI: 1.09-7.15; P = 0.03), especially when hCG was cultured with PBMCs for 48 h or PBMCs administration was performed two or three days before embryo transfer (ET). Neither the dosage of the hCG co-cultured with PBMCs nor the mean concentration of the administered PBMCs appeared to influence the therapeutic efficiency. In conclusion, intrauterine administration of PBMCs co-cultured with hCG for 48 h, conducted two or three days before ET, could be an effective therapy for women experiencing RIF. Due to the limitations of sample size and quality of the included studies, further high-quality studies with large sample sizes are warranted to optimize the parameters of hCG and PBMC co-culture to help more RIF patients benefit from this therapy.


Assuntos
Gonadotropina Coriônica/metabolismo , Implantação do Embrião/imunologia , Transferência Embrionária/métodos , Infertilidade Feminina/terapia , Leucócitos Mononucleares/transplante , Cultura Primária de Células/métodos , Células Cultivadas , Meios de Cultura/metabolismo , Feminino , Humanos , Infertilidade Feminina/imunologia , Leucócitos Mononucleares/imunologia , Gravidez , Taxa de Gravidez , Resultado do Tratamento , Útero/imunologia
10.
Clin Podiatr Med Surg ; 38(2): 235-244, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33745654

RESUMO

The treatment of Achilles tendinitis from conservative to minimally invasive to surgery gives patients a wide range of treatment options for this common pathology. The use and role of biologics to augment this treatment is emerging. The use of biologics may enhance the healing potential of the Achilles tendon when conservative treatment fails. There are a handful of biologics being investigated to obtain if improved outcomes can be maximized.


Assuntos
Tendão do Calcâneo , Tendinopatia/terapia , Transplante de Medula Óssea , Hepatócitos/transplante , Humanos , Leucócitos Mononucleares/transplante , Fator de Crescimento Derivado de Plaquetas/administração & dosagem , Plasma Rico em Plaquetas , Tecidos Suporte , Fator de Crescimento Transformador beta/administração & dosagem , Ultrassonografia de Intervenção , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Cicatrização
11.
Stem Cells Transl Med ; 10(6): 895-909, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33599112

RESUMO

The quality and quantity of endothelial progenitor cells (EPCs) are impaired in patients with diabetes mellitus patients, leading to reduced tissue repair during autologous EPC therapy. This study aimed to address the limitations of the previously described serum-free Quantity and Quality Control Culture System (QQc) using CD34+ cells by investigating the therapeutic potential of a novel mononuclear cell (MNC)-QQ. MNCs were isolated from 50 mL of peripheral blood of patients with diabetes mellitus and healthy volunteers (n = 13 each) and subjected to QQc for 7 days in serum-free expansion media with VEGF, Flt-3 ligand, TPO, IL-6, and SCF. The vascular regeneration capability of MNC-QQ cells pre- or post-QQc was evaluated with an EPC colony-forming assay, FACS, EPC culture, tube formation assay, and quantitative real time PCR. For in vivo assessment, 1 × 104 pre- and post-MNC-QQc cells from diabetic donors were injected into a murine wound-healing model using Balb/c nude mice. The percentage of wound closure and angio-vasculogenesis was then assessed. This study revealed vasculogenic, anti-inflammatory, and wound-healing effects of MNC-QQ therapy in both in vitro and in vivo models. This system addresses the low efficiency and efficacy of the current naïve MNC therapy for wound-healing in diabetic patients. As this technique requires a simple blood draw, isolation, and peripheral blood MNC suspension culture for only a week, it can be used as a simple and effective outpatient-based vascular and regenerative therapy for patients with diabetes mellitus.


Assuntos
Diabetes Mellitus , Leucócitos Mononucleares , Cicatrização , Animais , Meios de Cultura Livres de Soro , Humanos , Leucócitos Mononucleares/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Fisiológica
12.
J Immunol Methods ; 492: 113000, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33609532

RESUMO

BACKGROUND: Immunomodulation by mesenchymal stromal cells (MSCs) is a potentially important therapeutic modality. MSCs suppress peripheral blood mononuclear cell (PBMC) proliferation in vitro, suggesting a mechanism for suppressing inflammatory responses in vivo. This study details the interactions of PBMCs and MSCs. METHODS: Pooled human PBMCs and MSCs were co-cultured at different MSC:PBMC ratios and harvested from 0 to 120 h, with and without phytohaemagglutin A (PHA) stimulation. Proliferation of adherent MSCs and non-adherent PBMCs was assessed by quantitation of ATP levels. PBMC surface marker expression was analyzed by flow cytometry. Indoleamine 2,3-dioxygenase (IDO) activity was determined by kynurenine assay and IDO mRNA by RT-PCR. Cytokine release was measured by ELISA. Immunofluorescent microscopy detected MSC, PBMC, monocyte (CD14+) and apoptotic events. RESULTS: PBMC proliferation in response to PHA gave a robust ATP signal by 72 h, which was suppressed by co-culture with densely plated MSCs. Very low level MSC seeding densities relative to PBMC number reproducibly stimulated PBMC proliferation. The CD4+/CD3+ population significantly decreased over time while the CD8+/CD3+ population significantly increased. No change in CD4+/CD8+ ratio is seen with high density MSC co-culture; very low density MSCs augment the changes seen in PHA stimulated PBMCs alone. IDO activity in MSCs co-cultured with PBMCs correlated with PBMC suppression. MSCs increased the secretion of IL-10 and IL-6 from stimulated co-cultures and decreased TNF-α secretion. In stimulated co-culture, low density MSCs decreased in number; fluorescence immunomicroscopy detected association of PBMC with MSC and phosphatidyl serine externalization in both cell populations. CONCLUSIONS: A bidirectional interaction between MSCs and PBMCs occurs during co-culture. High numbers of MSCs inhibit PHA-stimulated PBMC proliferation and the PBMC response to stimulation; low numbers of MSCs augment these responses. Low density MSCs are susceptible to attrition, apparently by PBMC-induced apoptosis. These results may have direct application when considering therapeutic dosing of patients; low MSC doses may have unintended detrimental consequences.


Assuntos
Comunicação Celular/imunologia , Leucócitos Mononucleares/imunologia , Células-Tronco Mesenquimais/imunologia , Cultura Primária de Células/métodos , Proliferação de Células/efeitos dos fármacos , Transplante de Células , Técnicas de Cocultura/métodos , Humanos , Leucócitos Mononucleares/transplante , Mitógenos/farmacologia , Fito-Hemaglutininas/farmacologia
13.
Nat Commun ; 12(1): 135, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33420081

RESUMO

Since most variants that impact polygenic disease phenotypes localize to non-coding genomic regions, understanding the consequences of regulatory element variants will advance understanding of human disease mechanisms. Here, we report that the systemic lupus erythematosus (SLE) risk variant rs2431697 as likely causal for SLE through disruption of a regulatory element, modulating miR-146a expression. Using epigenomic analysis, genome-editing and 3D chromatin structure analysis, we show that rs2431697 tags a cell-type dependent distal enhancer specific for miR-146a that physically interacts with the miR-146a promoter. NF-kB binds the disease protective allele in a sequence-specific manner, increasing expression of this immunoregulatory microRNA. Finally, CRISPR activation-based modulation of this enhancer in the PBMCs of SLE patients attenuates type I interferon pathway activation by increasing miR-146a expression. Our work provides a strategy to define non-coding RNA functional regulatory elements using disease-associated variants and provides mechanistic links between autoimmune disease risk genetic variation and disease etiology.


Assuntos
Epigênese Genética/imunologia , Lúpus Eritematoso Sistêmico/genética , MicroRNAs/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Sistemas CRISPR-Cas , Estudos de Casos e Controles , Linhagem Celular Tumoral , Feminino , Predisposição Genética para Doença , Técnicas de Genotipagem , Células HEK293 , Voluntários Saudáveis , Humanos , Interferon Tipo I/metabolismo , Leucócitos Mononucleares/transplante , Lúpus Eritematoso Sistêmico/sangue , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Polimorfismo de Nucleotídeo Único , Cultura Primária de Células , Regiões Promotoras Genéticas , RNA-Seq , Transdução de Sinais/genética , Transdução de Sinais/imunologia
14.
Brain Res ; 1758: 147292, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33516814

RESUMO

Bone marrow mononuclear cells (BMMCs) have been identified as a relevant therapeutic strategy for the treatment of several chronic diseases of the central nervous system. The aim of this work was to evaluate whether intravenous treatment with BMMCs facilitates the reconnection of lesioned cortico-cortical and cortico-striatal pathways, together with motor recovery, in injured adult Wistar rats using an experimental model of unilateral focal neocortical ischaemia. Animals with cerebral cortex ischaemia underwent neural tract tracing for axonal fibre analysis, differential expression analysis of genes involved in apoptosis and neuroplasticity by RT-qPCR, and motor performance assessment by the cylinder test. Quantitative and qualitative analyses of axonal fibres labelled by an anterograde neural tract tracer were performed. Ischaemic animals treated with BMMCs showed a significant increase in axonal sprouting in the ipsilateral neocortex and in the striatum contralateral to the injured cortical areas compared to untreated rodents. In BMMC-treated animals, there was a trend towards upregulation of the Neurotrophin-3 gene compared to the other genes, as well as modulation of apoptosis by BMMCs. On the 56th day after ischaemia, BMMC-treated animals showed significant improvement in motor performance compared to untreated rats. These results suggest that in the acute phase of ischaemia, Neurotrophin-3 is upregulated in response to the lesion itself. In the long run, therapy with BMMCs causes axonal sprouting, reconnection of damaged neuronal circuitry and a significant increase in motor performance.


Assuntos
Transplante de Medula Óssea/métodos , Isquemia Encefálica/patologia , Leucócitos Mononucleares/transplante , Regeneração Nervosa/fisiologia , Neurotrofina 3/biossíntese , Recuperação de Função Fisiológica/fisiologia , Animais , Axônios/fisiologia , Masculino , Atividade Motora/fisiologia , Neocórtex , Ratos , Ratos Wistar , Regulação para Cima
15.
Cancer Sci ; 112(3): 1161-1172, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33410215

RESUMO

Activation of CD8+ Tax-specific CTL is a new therapeutic concept for adult T-cell leukemia (ATL) caused by HTLV-1. A recent clinical study of the dendritic cell vaccine pulsed with Tax peptides corresponding to CTL epitopes showed promising outcomes in ATL patients possessing limited human leukocyte antigen (HLA) alleles. In this study, we aimed to develop another immunotherapy to activate Tax-specific CTL without HLA limitation by using patients' own HTLV-1-infected cells as a vaccine. To examine the potential of HTLV-1-infected T-cells to activate CTL via antigen presenting cells, we established a unique co-culture system. We demonstrated that mitomycin C-treated HLA-A2-negative HTLV-1-infected T-cell lines or short-term cultured peripheral blood mononuclear cells (PBMC) derived from ATL patients induced cross-presentation of Tax antigen in co-cultured HLA-A2-positive antigen presenting cells, resulting in activation of HLA-A2-restricted CD8+ Tax-specific CTL. This effect was not inhibited by a reverse transcriptase inhibitor. IL-12 production and CD86 expression were also induced in antigen presenting cells co-cultured with HTLV-1-infected cells at various levels, which were improved by pre-treatment of the infected cells with histone deacetylase inhibitors. Furthermore, monocyte-derived dendritic cells induced from PBMC of a chronic ATL patient produced IL-12 and expressed enhanced levels of CD86 when co-cultured with autologous lymphocytes that had been isolated from the same PBMC and cultured for several days. These findings suggest that short-term cultured autologous PBMC from ATL patients could potentially serve as a vaccine to evoke Tax-specific CTL responses.


Assuntos
Vacinas Anticâncer/administração & dosagem , Técnicas de Cultura de Células , Infecções por HTLV-I/terapia , Imunoterapia/métodos , Leucemia-Linfoma de Células T do Adulto/terapia , Leucócitos Mononucleares/transplante , Antígenos Virais/imunologia , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Apresentação Cruzada/imunologia , Produtos do Gene tax/imunologia , Antígeno HLA-A2/imunologia , Antígeno HLA-A2/metabolismo , Infecções por HTLV-I/sangue , Infecções por HTLV-I/imunologia , Inibidores de Histona Desacetilases/farmacologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Humanos , Leucemia-Linfoma de Células T do Adulto/sangue , Leucemia-Linfoma de Células T do Adulto/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Mitomicina/farmacologia , Linfócitos T Citotóxicos/imunologia , Fatores de Tempo , Transplante Autólogo
16.
Int J Cancer ; 148(11): 2815-2824, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33368276

RESUMO

The canonical Wnt signaling pathway is activated in numerous contexts, including normal and cancerous tissues. Here, we describe a synthetic cell-based therapeutic strategy that inhibits aberrant Wnt activity in specific focuses without interfering with the normal tissues in vivo. As a proof of principle, we generated a triple transgenic zebrafish liver cancer model that conditionally expressed human MET and induced ectopic Wnt signaling in hepatocytes. Then, we generated a customized synthetic Notch receptor (synNotch) cascade to express Wnt inhibitor DKK1 in Jurkat T cells and human peripheral blood mononuclear cells (PBMCs) after recognizing MET as antigen. After that, the synNotch PBMCs were sorted and microinjected into different tissues of the zebrafish model. In MET-expressing cancerous liver tissues, the injected cells expressed DKK1 and inhibited the local proliferation and Wnt activity; while in the yolk sac without MET, the injected cells remained inactive. Overall, our studies revealed the use of synthetic cells with antigen receptors to improve the spatiotemporal accuracy of anti-Wnt therapy, and proposed that the genetically humanized zebrafish model may serve as a small-scale and highly optically accessible platform for the functional evaluation of human synthetic cells.


Assuntos
Proteínas de Ligação a DNA/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Leucócitos Mononucleares/transplante , Neoplasias Hepáticas Experimentais/terapia , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-myc/genética , Biologia Sintética/métodos , Proteínas de Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Proliferação de Células , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Jurkat , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Camundongos , Mutação , Estudo de Prova de Conceito , Proteínas Proto-Oncogênicas c-met/metabolismo , Via de Sinalização Wnt , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
17.
Stem Cells Transl Med ; 10(1): 14-26, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32902182

RESUMO

The aim of this study was to evaluate the safety and efficacy of autologous bone marrow mononuclear cell transplantation combined with educational intervention for children with autism spectrum disorder. An open-label clinical trial was performed from July 2017 to August 2019 at Vinmec International Hospital, Hanoi, Vietnam. Thirty children who fulfilled the autism criteria of the Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition, and had Childhood Autism Rating Scale (CARS) scores >37 were selected. Bone marrow was harvested by anterior iliac crest puncture under general anesthesia. The volume collected was as follows: 8 mL/kg for patients under 10 kg (80 mL + [body weight in kg - 10] × 7 mL) for patients above 10 kg. Mononuclear cells were isolated with a Ficoll gradient and then infused intrathecally. The same procedure was repeated 6 months later. After the first transplantation, all patients underwent 8 weeks of educational intervention based on the Early Start Denver Model. There were no severe adverse events associated with transplantation. The severity of autism spectrum disorder (ASD) was significantly reduced, with the median CARS score decreasing from 50 (range 40-55.5) to 46.5 (range 33.5-53.5) (P < .05). Adaptive capacity increased, with the median Vineland Adaptive Behavior Scales score rising from 53.5 to 60.5. Social communication, language, and daily skills improved markedly within 18 months after transplantation. Conversely, repetitive behaviors and hyperactivity decreased remarkably. Autologous bone marrow mononuclear cell transplantation in combination with behavioral intervention was safe and well tolerated in children with ASD (Trial registration: ClinicalTrials.gov identifier: NCT03225651).


Assuntos
Transtorno do Espectro Autista , Transplante de Medula Óssea , Leucócitos Mononucleares/transplante , Transtorno do Espectro Autista/terapia , Medula Óssea , Criança , Humanos , Vietnã
18.
Plast Reconstr Surg ; 147(2): 373-385, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33235046

RESUMO

BACKGROUND: Fat graft ischemia impedes us from having satisfying long-term results. The quality and quantity culture is a 1-week cell culture that increases the vasculogenic potential of peripheral blood mononuclear cells (PBMNC). This in vivo murine model investigates whether enrichment with quality and quantity-cultured human mononuclear cells (MNC-QQ) improves the vascularization in the human fat graft and whether this decreases the tissue loss. METHODS: Human adipose tissue, PBMNC, MNC-QQ, and stromal vascular fraction were prepared. First, PBMNC, MNC-QQ, and stromal vascular fraction were compared in vitro for vasculogenic potential by endothelial progenitor cell colony-forming and culture assays. Second, 0.25-g fat grafts were created with 1 × 106 PBMNC (n = 16), 1 × 106 MNC-QQ (n = 16), 1 × 106 stromal vascular fraction (n = 16), or phosphate-buffered saline as control (n = 16) before grafting in BALB/c nude mice. Grafts were analyzed for weight persistence, vessel formation by CD31 immunohistochemistry, and angiogenic markers by quantitative polymerase chain reaction. RESULTS: MNC-QQ develop more definitive endothelial progenitor cell colonies and more functional endothelial progenitor cells compared to PBMNC and stromal vascular fraction. Weight persistence after 7 weeks was significantly higher in grafts with MNC-QQ (89.8 ± 3.5 percent) or stromal vascular fraction (90.1 ± 4.2 percent) compared with control (70.4 ± 6.3 percent; p < 0.05). MNC-QQ-enriched grafts had the highest vessel density (96.6 ± 6.5 vessels/mm2; control, 70.4 ± 5.6 vessels/mm2; p < 0.05). MNC-QQ exerted a direct vasculogenic effect through vascular integration and a potential paracrine vascular endothelial growth factor-mediated effect. CONCLUSION: Quality and quantity-cultured human mononuclear cells containing endothelial progenitor cells stimulate fat graft vascularization and enhance graft survival in a rodent recipient.


Assuntos
Tecido Adiposo/transplante , Sobrevivência de Enxerto/fisiologia , Leucócitos Mononucleares/transplante , Neovascularização Fisiológica , Adipócitos/fisiologia , Tecido Adiposo/citologia , Animais , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Modelos Animais de Doenças , Células Progenitoras Endoteliais/fisiologia , Feminino , Humanos , Leucócitos Mononucleares/fisiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Cultura Primária de Células , Células Estromais/transplante
19.
PLoS One ; 15(12): e0244160, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33370374

RESUMO

Ischemia-reperfusion injury is an important contributor to acute kidney injury and a major factor affecting early functional recovery after kidney transplantation. We conducted this experiment to investigate the protective effect of induced multipotent stem cell transplantation on renal ischemia-reperfusion injury. Forty rabbits were divided into four groups of 10 rabbits each. Thirty rabbits were used to establish the renal ischemia-reperfusion injury model, and ten rabbits served as the model group and were not treated. Among the 30 rabbits with renal ischemia-reperfusion injury, 10 rabbits were treated with induced peripheral blood mononuclear cells (PBMCs), and 10 other rabbits were treated with noninduced PBMCs. After three weekly treatments, the serum creatinine levels, urea nitrogen levels and urine protein concentrations were quantified. The kidneys were stained with hematoxylin-eosin (HE), periodic acid-Schiff (PAS) and Masson's trichrome and then sent for commercial metabolomic testing. The kidneys of the rabbits in the model group showed different degrees of pathological changes, and the recovery of renal function was observed in the group treated with induced cells. The results indicate that PBMCs differentiate into multipotent stem cells after induction and exert a therapeutic effect on renal ischemia-reperfusion injury.


Assuntos
Clara de Ovo/química , Rim/irrigação sanguínea , Leucócitos Mononucleares/transplante , Traumatismo por Reperfusão/terapia , Animais , Diferenciação Celular , Extratos Celulares/farmacologia , Células Cultivadas , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Coelhos
20.
Int J Mol Sci ; 21(17)2020 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-32867222

RESUMO

Stroke remains a major cause of serious disability because the brain has a limited capacity to regenerate. In the last two decades, therapies for stroke have dramatically changed. However, half of the patients cannot achieve functional independence after treatment. Presently, cell-based therapies are being investigated to improve functional outcomes. This review aims to describe conventional cell therapies under clinical trial and outline the novel concept of polarized cell therapies based on protective cell phenotypes, which are currently in pre-clinical studies, to facilitate functional recovery after post-reperfusion treatment in patients with ischemic stroke. In particular, non-neuronal stem cells, such as bone marrow-derived mesenchymal stem/stromal cells and mononuclear cells, confer no risk of tumorigenesis and are safe because they do not induce rejection and allergy; they also pose no ethical issues. Therefore, recent studies have focused on them as a cell source for cell therapies. Some clinical trials have shown beneficial therapeutic effects of bone marrow-derived cells in this regard, whereas others have shown no such effects. Therefore, more clinical trials must be performed to reach a conclusion. Polarized microglia or peripheral blood mononuclear cells might provide promising therapeutic strategies after stroke because they have pleiotropic effects. In traumatic injuries and neurodegenerative diseases, astrocytes, neutrophils, and T cells were polarized to the protective phenotype in pre-clinical studies. As such, they might be useful therapeutic targets. Polarized cell therapies are gaining attention in the treatment of stroke and neurological diseases.


Assuntos
AVC Isquêmico/terapia , Leucócitos Mononucleares/transplante , Transplante de Células-Tronco Mesenquimais/métodos , Doenças do Sistema Nervoso/terapia , Animais , Polaridade Celular , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Humanos , AVC Isquêmico/fisiopatologia , Leucócitos Mononucleares/citologia , Células-Tronco Mesenquimais/citologia , Microglia/citologia , Microglia/transplante , Doenças do Sistema Nervoso/fisiopatologia , Recuperação de Função Fisiológica , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...