Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
JCI Insight ; 8(20)2023 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-37698919

RESUMO

Retinoic acid receptor (RAR) signaling is essential for mammalian kidney development but, in the adult kidney, is restricted to occasional collecting duct epithelial cells. We now show that there is widespread reactivation of RAR signaling in proximal tubular epithelial cells (PTECs) in human sepsis-associated acute kidney injury (AKI) and in mouse models of AKI. Genetic inhibition of RAR signaling in PTECs protected against experimental AKI but was unexpectedly associated with increased expression of the PTEC injury marker Kim1. However, the protective effects of inhibiting PTEC RAR signaling were associated with increased Kim1-dependent apoptotic cell clearance, or efferocytosis, and this was associated with dedifferentiation, proliferation, and metabolic reprogramming of PTECs. These data demonstrate the functional role that reactivation of RAR signaling plays in regulating PTEC differentiation and function in human and experimental AKI.


Assuntos
Injúria Renal Aguda , Túbulos Renais Proximais , Camundongos , Animais , Humanos , Túbulos Renais Proximais/metabolismo , Tretinoína/farmacologia , Tretinoína/metabolismo , Rim/metabolismo , Injúria Renal Aguda/metabolismo , Células Epiteliais/metabolismo , Mamíferos
3.
Am J Physiol Renal Physiol ; 323(4): F479-F491, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35979965

RESUMO

Kidney organoids derived from human or rodent pluripotent stem cells have glomerular structures and differentiated/polarized nephron segments. Although there is an increasing understanding of the patterns of expression of transcripts and proteins within kidney organoids, there is a paucity of data regarding functional protein expression, in particular on transporters that mediate the vectorial transport of solutes. Using cells derived from kidney organoids, we examined the functional expression of key ion channels that are expressed in distal nephron segments: the large-conductance Ca2+-activated K+ (BKCa) channel, the renal outer medullary K+ (ROMK, Kir1.1) channel, and the epithelial Na+ channel (ENaC). RNA-sequencing analyses showed that genes encoding the pore-forming subunits of these transporters, and for BKCa channels, key accessory subunits, are expressed in kidney organoids. Expression and localization of selected ion channels was confirmed by immunofluorescence microscopy and immunoblot analysis. Electrophysiological analysis showed that BKCa and ROMK channels are expressed in different cell populations. These two cell populations also expressed other unidentified Ba2+-sensitive K+ channels. BKCa expression was confirmed at a single channel level, based on its high conductance and voltage dependence of activation. We also found a population of cells expressing amiloride-sensitive ENaC currents. In summary, our results show that human kidney organoids functionally produce key distal nephron K+ and Na+ channels.NEW & NOTEWORTHY Our results show that human kidney organoids express key K+ and Na+ channels that are expressed on the apical membranes of cells in the aldosterone-sensitive distal nephron, including the large-conductance Ca2+-activated K+ channel, renal outer medullary K+ channel, and epithelial Na+ channel.


Assuntos
Células-Tronco Pluripotentes Induzidas , Canais de Potássio Corretores do Fluxo de Internalização , Aldosterona/metabolismo , Amilorida/farmacologia , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Rim/metabolismo , Organoides/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , RNA/metabolismo , Sódio/metabolismo
4.
Stem Cell Res Ther ; 13(1): 76, 2022 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-35189973

RESUMO

BACKGROUND: Hemolysis occurs in many injury settings and can trigger disease processes. In the kidney, extracellular hemoglobin can induce damage via several mechanisms. These include oxidative stress, mitochondrial dysfunction, and inflammation, which promote fibrosis and chronic kidney disease. Understanding the pathophysiology of these injury pathways offers opportunities to develop new therapeutic strategies. METHODS: To model hemolysis-induced kidney injury, human kidney organoids were treated with hemin, an iron-containing porphyrin, that generates reactive oxygen species. In addition, we developed an induced pluripotent stem cell line expressing the biosensor, CytochromeC-GFP (CytoC-GFP), which provides a real-time readout of mitochondrial morphology, health, and early apoptotic events. RESULTS: We found that hemin-treated kidney organoids show oxidative damage, increased expression of injury markers, impaired functionality of organic anion and cation transport and undergo fibrosis. Injury could be detected in live CytoC-GFP organoids by cytoplasmic localization of fluorescence. Finally, we show that 4-(phenylthio)butanoic acid, an HDAC inhibitor with anti-fibrotic effects in vivo, reduces hemin-induced human kidney organoid fibrosis. CONCLUSION: This work establishes a hemin-induced model of kidney organoid injury. This platform provides a new tool to study the injury and repair response pathways in human kidney tissue and will assist in the development of new therapeutics.


Assuntos
Células-Tronco Pluripotentes , Insuficiência Renal Crônica , Humanos , Rim/metabolismo , Organoides/metabolismo , Estresse Oxidativo , Insuficiência Renal Crônica/metabolismo
5.
Asia Pac J Clin Oncol ; 18(1): 44-51, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32970928

RESUMO

AIMS: The purpose of this study was to design and manufacture CD19 chimeric antigen receptor (CAR)-modified T cells for clinical use in Thailand, as a model for how this technology can be directly applied at individual institutions treating high-risk leukemia patients. METHODS: We constructed second-generation CAR T cells expressing CD19 scFV-CD28-CD3ζ with different lengths of the spacer region: full, intermediate, and short length, by using a lentiviral vector. We wanted to determine whether the difference in length of the spacer would affect the cytotoxic potential of the CD19 CAR T cells against the leukemic cells. RESULTS: We found that all constructs of CD19 CAR T cells exhibited a similar level of cytotoxicity against several human lymphoma and leukemia cell lines. For the clinical application, we chose the intermediate length spacer construct CD19 CAR T cells, hypothesizing that the highest transduction efficiency coupled with a slower initial proliferation in vitro might lead to effective leukemic cell kill, yet a lower probability for serious clinical side effects. We then tested the clinical efficacy of our CD19 CAR T cells in one patient with refractory/relapsed acute B-cell lymphoblastic leukemia. This patient indeed had minimal clinical side effects after the CAR T-cell infusion, and he remains in an unmaintained, ongoing complete remission 10+ months after his T-cell treatment. CONCLUSION: Our CD19 CAR T cells demonstrated efficacies in acute lymphoblastic B-cell leukemia, and will be used to establish an immunotherapeutic program for high-risk B-cell acute lymphoblastic leukemia in Thailand. We propose that this approach can be used as a model for how this new exciting technology can be applied directly at individual institutions that treat (a large number of) patients with high-risk leukemia.


Assuntos
Antígenos CD19 , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Imunoterapia Adotiva , Masculino , Indução de Remissão , Linfócitos T
6.
Cancers (Basel) ; 13(15)2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34359679

RESUMO

Circulating atypical cells (CAC) are released from a primary tumour site into peripheral blood and are indicators of cancer metastasis. CAC occur at very low frequency in circulating blood, and their detection remains challenging. Moreover, white blood cells (WBC) are the major contaminant in enriched CAC samples. Here, we developed matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) as a novel CAC characterization platform. Main spectra profiles (MSP) of normal and cancer cells were generated by MALDI-TOF MS, and a cell-main spectra database was then compiled and analysed using the MALDI Biotyper software. Logarithmic scores accurately predicted distinct cell types. The feasibility of this workflow was then validated using simulated samples, which were prepared by 5000 WBC of three healthy individuals spiked with varying numbers (3, 6, 12, 25, 50, and 100) of lung, colon, or prostate cancer cells. MALDI-TOF MS was able to detect cancer cells down to six cells over the background noise of 5000 WBC with significantly higher predictive scores as compared to WBC alone. Further development of cell-MSP database to cover all cancer types sourced from cell lines and patient tumours may enable the use of MALDI-TOF MS as a cancer-screening platform in clinical settings in the future.

7.
Biomedicines ; 9(8)2021 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-34440185

RESUMO

Chemotherapy in childhood leukemia is associated with late morbidity in leukemic survivors, while certain patient subsets are relatively resistant to standard chemotherapy. It is therefore important to identify new agents with sensitivity and selectivity towards leukemic cells, while having less systemic toxicity. Peptide-based therapeutics has gained a great deal of attention during the last few years. Here, we used an integrative workflow combining mass spectrometric peptide library construction, in silico anticancer peptide screening, and in vitro leukemic cell studies to discover a novel anti-leukemic peptide having 3+ charges and an alpha helical structure, namely HMP-S7, from human breast milk. HMP-S7 showed cytotoxic activity against four distinct leukemic cell lines in a dose-dependent manner but had no effect on solid malignancies or representative normal cells. HMP-S7 induced leukemic cell death by penetrating the plasma membrane to enter the cytoplasm and cause the leakage of lactate dehydrogenase, thus acting in a membranolytic manner. Importantly, HMP-S7 exhibited anti-leukemic effects against patient-derived leukemic cells ex vivo. In conclusion, HMP-S7 is a selective anti-leukemic peptide with promise, which requires further validation in preclinical and clinical studies.

8.
Am J Physiol Renal Physiol ; 318(4): F971-F978, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32150447

RESUMO

Acute kidney injury (AKI) remains a major global healthcare problem, and there is a need to develop human-based models to study AKI in vitro. Toward this goal, we have characterized induced pluripotent stem cell-derived human kidney organoids and their response to cisplatin, a chemotherapeutic drug that induces AKI and preferentially damages the proximal tubule. We found that a single treatment with 50 µM cisplatin induces hepatitis A virus cellular receptor 1 (HAVCR1) and C-X-C motif chemokine ligand 8 (CXCL8) expression, DNA damage (γH2AX), and cell death in the organoids but greatly impairs organoid viability. DNA damage was not specific to the proximal tubule but also affected the distal tubule and interstitial cell populations. This lack of specificity correlated with low expression of proximal tubule-specific SLC22A2/organic cation transporter 2 (OCT2) for cisplatin. To improve viability, we developed a repeated low-dose regimen of 4 × 5 µM cisplatin over 7 days and found this caused less toxicity while still inducing a robust injury response that included secretion of known AKI biomarkers and inflammatory cytokines. This work validates the use of human kidney organoids to model aspects of cisplatin-induced injury, with the potential to identify new AKI biomarkers and develop better therapies.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Antineoplásicos/toxicidade , Cisplatino/toxicidade , Dano ao DNA , Túbulos Renais Proximais/efeitos dos fármacos , Organoides/efeitos dos fármacos , Injúria Renal Aguda/genética , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Antineoplásicos/metabolismo , Células Cultivadas , Cisplatino/metabolismo , Relação Dose-Resposta a Droga , Receptor Celular 1 do Vírus da Hepatite A/genética , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Histonas/metabolismo , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Transportador 2 de Cátion Orgânico/metabolismo , Organoides/metabolismo , Organoides/patologia , Fatores de Tempo
9.
J Immunother ; 42(4): 110-118, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30921263

RESUMO

The third-party umbilical cord blood (UCB)-derived regulatory T cells (Treg) are an alternative to donor-derived Treg as cellular therapy of graft-versus-host disease following hematopoietic stem cell transplantation. However, their suppressive characteristics against autologous and allogeneic T effector cells (Teff) have rarely been documented. The exact role of UCB-Treg in hematologic malignancies is also uncertain. Here, we investigated the direct effects of UCB-Treg on the proliferation of autologous Teff, as compared with allogeneic Teff, and also determined cellular fates of lymphoblasts after UCB-Treg co-culture. UCB-Treg were isolated from 8 UCB samples using 2-step immunomagnetic bead sorting. After 10-day ex vivo expansion, up to 60-fold increase in cell number with 76.7%±4.9% of CD4CD25CD127FoxP UCB-Treg was obtained. Further characterization showed that ex vivo-expanded UCB-Treg contained a higher proportion of CD95CD45RACCR4Treg-B subpopulation compared with the CD95CD45RACCR4Treg-A subpopulation (13.0%±4.8% vs. 0.8%±0.7%; P<0.05), along with the detecting of substantial amounts of secretory IL-10 (57.7±17.8 pg/mL) and TGF-ß1 (196.5±29.7 pg/mL) in culture supernatants. After 4 days co-culture with UCB-Treg (at the ratio of 1:1), the proliferation of autologous and allogeneic Teff was decreased comparably (43.6%±17.5% vs. 37.6±17.7%; P=0.437). Suppression was independent of HLA-A, B, and DRB1 compatibility between UCB-Treg and Teff. UCB-Treg co-culture with various lymphoblasts showed proliferative suppression of Jurkat T lymphoblasts (45.4%±20.5% at the ratio of 1:1), but not Namalwa and Raji B lymphoblasts. All lymphoblasts had no significant cell apoptosis or death after co-culture. In conclusion, the ex vivo-expanded UCB-Treg had no difference in autologous and allogeneic Teff suppression. UCB-Treg therapy in patients with graft-versus-host disease who have a primary disease of T-cell leukemia may have additional benefits in the prevention of relapsed disease.


Assuntos
Transferência Adotiva , Terapia Baseada em Transplante de Células e Tecidos , Sangue Fetal/citologia , Tolerância Imunológica , Imunomodulação , Linfócitos T Reguladores/imunologia , Aloenxertos , Apoptose/imunologia , Comunicação Celular/imunologia , Técnicas de Cultura de Células , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células Cultivadas , Xenoenxertos , Humanos , Imunofenotipagem , Ativação Linfocitária/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo
10.
Biomed Res Int ; 2018: 1812535, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29682521

RESUMO

Neuroblastoma (NB) is the most common extracranial solid tumor in childhood with 5-year survival rate of 40% in high-risk patients despite intensive therapies. Recently, adoptive cell therapy, particularly chimeric antigen receptor (CAR) T cell therapy, represents a revolutionary treatment for hematological malignancies. However, there are challenges for this therapeutic strategy with solid tumors, as a result of the immunosuppressive nature of the tumor microenvironment (TME). Cancer cells have evolved multiple mechanisms to escape immune recognition or to modulate immune cell function. Several subtypes of immune cells that infiltrate tumors can foster tumor development, harbor immunosuppressive activity, and decrease an efficacy of adoptive cell therapies. Therefore, an understanding of the dual role of the immune system under the influences of the TME has been crucial for the development of effective therapeutic strategies against solid cancers. This review aims to depict key immune players and cellular pathways involved in the dynamic interplay between the TME and the immune system and also to address challenges and prospective development of adoptive T cell transfer for neuroblastoma.


Assuntos
Neuroblastoma/imunologia , Neuroblastoma/terapia , Transferência Adotiva/métodos , Animais , Humanos , Imunoterapia Adotiva/métodos , Receptores de Antígenos de Linfócitos T/imunologia , Linfócitos T/imunologia , Microambiente Tumoral/imunologia
11.
J Oncol ; 2018: 7946021, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30643519

RESUMO

Neuroblastoma (NB) is the most common extracranial tumor of childhood with poor prognosis in a high-risk group. An obstacle in the development of treatment for solid tumors is the immunosuppressive nature of the tumor microenvironment (TME). Regulatory T cells (Tregs) represent a T cell subset with specialized function in immune suppression and maintaining self-tolerance. Tregs resident within the tumor milieu is believed to play an important role in immune escape mechanisms. The role of the NB microenvironment in promoting Treg phenotype has never been elucidated. Herein, we demonstrated that the NB microenvironment promoted T cell activation and one NB cell line, SK-N-SH, manifested an ability to induce Treg differentiation. We identified tumor-derived HMGB1 as a potential protein responsible for Treg phenotype induction. By neutralizing HMGB1, Treg differentiation was abolished. Finally, we adopted a dataset of 498 pediatric NB via the NCBI GEO database, accession GSE49711, to validate clinical relevance of HMGB1 overexpression. Up to 11% of patients had HMGB1-overexpressed tumors. Moreover, this patient subpopulation showed higher risks of tumor progression, relapse, or death. Our findings emphasize the importance of immunological signature of tumor cells for appropriate therapeutic approach. Upregulation of secretory HMGB1 may contribute to suppression of antitumor immunity through induction of Tregs in the NB microenvironment.

12.
Mol Med Rep ; 12(3): 3265-3272, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018028

RESUMO

Hypoxia is associated with tumor progression and poor prognosis in several cancer types. The present study aimed to examine the contribution of hypoxia (1% O2) to cancer progression in a cholangiocarcinoma cell line, RMCCA­1. The molecular basis of the hypoxic response pathway was investigated. The results showed that hypoxia significantly accelerated cancer cell proliferation and enhanced cell invasion (P<0.05). By using receptor tyrosine kinase and intracellular signaling antibody array kits, an increased phosphorylation/activation of a number of signaling molecules, particularly hepatocyte growth factor receptor (Met) and extracellular signal­regulated kinase (ERK) 1/2, was identified. Inhibition of Met and ERK by small hairpin RNA and U0126, respectively, significantly inhibited hypoxia­induced the invasive potential of RMCCA­1 cells (P<0.05). However, according to immunohistochemical analysis, hypoxia­inducible factor­1α expression was not correlated with cancer staging or tumor differentiation in 44 samples of cholangicarcinoma cases. The findings of the present study emphasized the importance of Met/ERK pathway activation as a key molecular event that may be responsible for a more invasive phenotype in hypoxic tumors and suggest Met as a potential target for the treatment of cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/complicações , Ductos Biliares/patologia , Colangiocarcinoma/complicações , Hipóxia/complicações , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-met/metabolismo , Neoplasias dos Ductos Biliares/metabolismo , Neoplasias dos Ductos Biliares/patologia , Ductos Biliares/metabolismo , Linhagem Celular Tumoral , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Invasividade Neoplásica/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...