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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Biomed Eng ; 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38424088

RESUMO

Kidney disease is a global health crisis affecting more than 850 million people worldwide. In the United States, annual Medicare expenditures for kidney disease and organ failure exceed $81 billion. Efforts to develop targeted therapeutics are limited by a poor understanding of the molecular mechanisms underlying human kidney disease onset and progression. Additionally, 90% of drug candidates fail in human clinical trials, often due to toxicity and efficacy not accurately predicted in animal models. The advent of ex vivo kidney models, such as those engineered from induced pluripotent stem (iPS) cells and organ-on-a-chip (organ-chip) systems have garnered considerable interest owing to their ability to more accurately model tissue development and patient-specific responses and drug toxicity. This review describes recent advances in developing kidney organoids and organ-chips by harnessing iPS cell biology to model human-specific kidney functions and disease states. We also discuss challenges that must be overcome to realize the potential of organoids and organ-chips as dynamic and functional conduits of the human kidney. Achieving these technological advances could revolutionize personalized medicine applications and therapeutic discovery for kidney disease. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 26 is May 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.

2.
Biomed Mater ; 17(6)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36044885

RESUMO

Nanoconfinement within flexible interfaces is a key step towards exploiting confinement effects in several biological and technological systems wherein flexible 2D materials are frequently utilized but are arduous to prepare. Hitherto unreported, the synthesis of 2D hydrogel nanosheets (HNSs) using a template- and catalyst-free process is developed representing a fertile ground for fundamental structure-property investigations. In due course of time, nucleating folds propagating along the edges trigger co-operative deformations of HNS generating regions of nanoconfinement within trapped water islands. These severely constricting surfaces force water molecules to pack within the nanoscale regime of HNS almost parallel to the surface bringing about phase transition into puckered rhombic ice with AA and AB Bernal stacking pattern, which was mostly restricted to molecular dynamics studies so far. Interestingly, under high lateral pressure and spatial inhomogeneity within nanoscale confinement, bilayer rhombic ice structures were formed with an in-plane lattice spacing of 0.31 nm. In this work, a systematic exploration of rhombic ice formation within HNS has been delineated using high-resolution transmission electron microscopy, and its ultrathin morphology was examined using atomic force microscopy. Scanning electron microscopy images revealed high porosity while mechanical testing presented young's modulus of 155 kPa with ∼84% deformation, whereas contact angle suggested high hydrophilicity. The combinations of nanosheets, porosity, nanoconfinement, hydrophilicity, and mechanical strength, motivated us to explore their application as a scaffold for cartilage regeneration, by inducing chondrogenesis of human Wharton Jelly derived mesenchymal stem cells. HNS promoted the formation of cell aggregates giving higher number of spheroid formation and a marked expression of chondrogenic markers (ColI, ColII, ColX, ACAN and S-100), thereby providing some cues for guiding chondrogenic differentiation.


Assuntos
Células-Tronco Mesenquimais , Geleia de Wharton , Diferenciação Celular , Células Cultivadas , Condrogênese , Humanos , Hidrogéis/química , Gelo
3.
J Sci Food Agric ; 102(14): 6491-6501, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35567417

RESUMO

BACKGROUND: Soil-dwelling human pathogens like Salmonella are transmitted by fresh produce such as tomato, spinach, onion and cabbage. With >2600 serovars, it is difficult to classify the good plant colonizers from the non-colonizers. Generally, soil microbiota are classified as autochthonous or zymogenous organisms, based on their ability to survive in soil. However, such information for soil-dwelling human pathogens is not available Thus there is a need to classify these organisms for designing a strategy to prevent their outbreak. Moreover, soil harbours a plethora of microbes, which can be screened for competitive organisms to control such human pathogens. RESULTS: In this study, we examined whether the morphotype based on the attachment factors (e.g., cellulose and curli fimbri) of Salmonella was important for its colonization of roots. Secondly, we tracked the location of the bacteria in the plant cell. Interestingly, most of the epidermal cells occupied by Salmonella showed propidium iodide-positive nuclei. As an extension of the study, a screening of competitive rhizospheric bacteria was performed. One isolate, identified as Lysinibacillus macroides, was able to inhibit the biofilm of Salmonella and subsequently reduced its colonization on roots. CONCLUSION: Based on this study, we classified the Rdar (red, dry and rough) morphotypes as good plant colonists. The ability to colonize and subsequent kill the live plant cell throws light on the zymogenous life cycle of soil-dwelling Salmonella. Additionally, Lysinibacillus macroides served as a biocontrol agent by reducing the burden of Salmonella in various vegetables. Such organisms can further be explored to prevent contamination of the food chain. © 2022 Society of Chemical Industry.


Assuntos
Celulose , Salmonella , Bacillaceae , Celulose/metabolismo , Humanos , Propídio , Solo
4.
Front Cell Dev Biol ; 10: 855340, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35517495

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the Coronavirus disease 2019 (COVID-19), which has resulted in over 5.9 million deaths worldwide. While cells in the respiratory system are the initial target of SARS-CoV-2, there is mounting evidence that COVID-19 is a multi-organ disease. Still, the direct affinity of SARS-CoV-2 for cells in other organs such as the kidneys, which are often targeted in severe COVID-19, remains poorly understood. We employed a human induced pluripotent stem (iPS) cell-derived model to investigate the affinity of SARS-CoV-2 for kidney glomerular podocytes, and examined the expression of host factors for binding and processing of the virus. We studied cellular uptake of the live SARS-CoV-2 virus as well as a pseudotyped virus. Infection of podocytes with live SARS-CoV-2 or spike-pseudotyped lentiviral particles revealed cellular uptake even at low multiplicity of infection (MOI) of 0.01. We found that direct infection of human iPS cell-derived podocytes by SARS-CoV-2 virus can cause cell death and podocyte foot process retraction, a hallmark of podocytopathies and progressive glomerular diseases including collapsing glomerulopathy observed in patients with severe COVID-19 disease. We identified BSG/CD147 and ACE2 receptors as key mediators of spike binding activity in human iPS cell-derived podocytes. These results show that SARS-CoV-2 can infect kidney glomerular podocytes in vitro via multiple binding interactions and partners, which may underlie the high affinity of SARS-CoV-2 for kidney tissues. This stem cell-derived model is potentially useful for kidney-specific antiviral drug screening and mechanistic studies of COVID-19 organotropism.

5.
Bioengineering (Basel) ; 9(5)2022 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-35621466

RESUMO

Podocytes derived from human induced pluripotent stem (hiPS) cells are enabling studies of kidney development and disease. However, many of these studies are carried out in traditional tissue culture plates that do not accurately recapitulate the molecular and mechanical features necessary for modeling tissue- and organ-level functionalities. Overcoming these limitations requires the design and application of tunable biomaterial scaffolds. Silk fibroin is an attractive biomaterial due to its biocompatibility and versatility, which include its ability to form hydrogels, sponge-like scaffolds, and electrospun fibers and membranes appropriate for tissue engineering and biomedical applications. In this study, we show that hiPS cells can be differentiated into post-mitotic kidney glomerular podocytes on electrospun silk fibroin membranes functionalized with laminin. The resulting podocytes remain viable and express high levels of podocyte-specific markers consistent with the mature cellular phenotype. The resulting podocytes were propagated for at least two weeks, enabling secondary cell-based applications and analyses. This study demonstrates for the first time that electrospun silk fibroin membrane can serve as a supportive biocompatible platform for human podocyte differentiation and propagation. We anticipate that the results of this study will pave the way for the use of electrospun membranes and other biomimetic scaffolds for kidney tissue engineering, including the development of co-culture systems and organs-on-chips microphysiological devices.

6.
ACS Chem Biol ; 17(12): 3341-3351, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34890187

RESUMO

The most severe forms of kidney diseases are often associated with irreversible damage to the glomerular podocytes, the highly specialized epithelial cells that encase glomerular capillaries and regulate the removal of toxins and waste from the blood. Several studies revealed significant changes to podocyte cytoskeletal structure during disease onset, suggesting possible roles of cellular mechanosensing in podocyte responses to injury. Still, this topic remains underexplored partly due to the lack of appropriate in vitro models that closely recapitulate human podocyte biology. Here, we leveraged our previously established method for the derivation of mature podocytes from human induced pluripotent stem cells (hiPSCs) to help uncover the roles of yes-associated protein (YAP), a transcriptional coactivator and mechanosensor, in podocyte injury response. We found that while the total expression levels of YAP remain relatively unchanged during Adriamycin (ADR)-induced podocyte injury, the YAP target genes connective tissue growth factor (CTGF) and cysteine-rich angiogenic inducer 61 (Cyr61) are significantly downregulated. Intriguingly, TEAD1 is significantly downregulated in podocytes injured with ADR. By examining multiple independent modes of cellular injury, we found that CTGF and Cyr61 expression are downregulated only when podocytes were exposed to molecules known to disrupt the cell's mechanical integrity or cytoskeletal structure. To our knowledge, this is the first report that the YAP-TEAD1 signaling axis is disrupted when stem cell-derived human podocytes experience biomechanical injury. Together, these results could help improve the understanding of kidney disease mechanisms and highlight CTGF and Cyr61 as potential therapeutic targets or biomarkers for patient stratification.


Assuntos
Células-Tronco Pluripotentes Induzidas , Podócitos , Humanos , Podócitos/metabolismo , Doxorrubicina/farmacologia , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Transdução de Sinais , Fatores de Transcrição de Domínio TEA
7.
bioRxiv ; 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34816259

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the Coronavirus disease 2019 (COVID-19), which was declared a pandemic by the World Health Organization (WHO) in March 2020. The disease has caused more than 5.1 million deaths worldwide. While cells in the respiratory system are frequently the initial target for SARS-CoV-2, clinical studies suggest that COVID-19 can become a multi-organ disease in the most severe cases. Still, the direct affinity of SARS-CoV-2 for cells in other organs such as the kidneys, which are often affected in severe COVID-19, remains poorly understood. METHOD: In this study, we employed a human induced pluripotent stem (iPS) cell-derived model to investigate the affinity of SARS-CoV-2 for kidney glomerular podocytes. We studied uptake of the live SARS-CoV-2 virus as well as pseudotyped viral particles by human iPS cell derived podocytes using qPCR, western blot, and immunofluorescence. Global gene expression and qPCR analyses revealed that human iPS cell-derived podocytes express many host factor genes (including ACE2, BSG/CD147, PLS3, ACTR3, DOCK7, TMPRSS2, CTSL CD209, and CD33) associated with SARS-CoV-2 binding and viral processing. RESULT: Infection of podocytes with live SARS-CoV-2 or spike-pseudotyped lentiviral particles revealed viral uptake by the cells at low Multiplicity of Infection (MOI of 0.01) as confirmed by RNA quantification and immunofluorescence studies. Our results also indicate that direct infection of human iPS cell-derived podocytes by SARS-CoV-2 virus can cause cell death and podocyte foot process retraction, a hallmark of podocytopathies and progressive glomerular diseases including collapsing glomerulopathy observed in patients with severe COVID-19 disease. Additionally, antibody blocking experiments identified BSG/CD147 and ACE2 receptors as key mediators of spike binding activity in human iPS cell-derived podocytes. CONCLUSION: These results show that SARS-CoV-2 can infect kidney glomerular podocytes in vitro . These results also show that the uptake of SARS-CoV-2 by kidney podocytes occurs via multiple binding interactions and partners, which may underlie the high affinity of SARS-CoV-2 for kidney tissues. This stem cell-derived model is potentially useful for kidney-specific antiviral drug screening and mechanistic studies of COVID-19 organotropism. SIGNIFICANT STATEMENT: Many patients with COVID19 disease exhibit multiorgan complications, suggesting that SARS-CoV-2 infection can extend beyond the respiratory system. Acute kidney injury is a common COVID-19 complication contributing to increased morbidity and mortality. Still, SARS-Cov-2 affinity for specialized kidney cells remain less clear. By leveraging our protocol for stem cell differentiation, we show that SARS-CoV-2 can directly infect kidney glomerular podocytes by using multiple Spike-binding proteins including ACE2 and BSG/CD147. Our results also indicate that infection by SARS-CoV-2 virus can cause podocyte cell death and foot process effacement, a hallmark of podocytopathies including collapsing glomerulopathy observed in patients with severe COVID-19 disease. This stem cell-derived model is potentially useful for kidney-specific antiviral drug screening and mechanistic studies of COVID-19 organotropism.

8.
Micromachines (Basel) ; 12(8)2021 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-34442589

RESUMO

Progress in understanding kidney disease mechanisms and the development of targeted therapeutics have been limited by the lack of functional in vitro models that can closely recapitulate human physiological responses. Organ Chip (or organ-on-a-chip) microfluidic devices provide unique opportunities to overcome some of these challenges given their ability to model the structure and function of tissues and organs in vitro. Previously established organ chip models typically consist of heterogenous cell populations sourced from multiple donors, limiting their applications in patient-specific disease modeling and personalized medicine. In this study, we engineered a personalized glomerulus chip system reconstituted from human induced pluripotent stem (iPS) cell-derived vascular endothelial cells (ECs) and podocytes from a single patient. Our stem cell-derived kidney glomerulus chip successfully mimics the structure and some essential functions of the glomerular filtration barrier. We further modeled glomerular injury in our tissue chips by administering a clinically relevant dose of the chemotherapy drug Adriamycin. The drug disrupts the structural integrity of the endothelium and the podocyte tissue layers, leading to significant albuminuria as observed in patients with glomerulopathies. We anticipate that the personalized glomerulus chip model established in this report could help advance future studies of kidney disease mechanisms and the discovery of personalized therapies. Given the remarkable ability of human iPS cells to differentiate into almost any cell type, this work also provides a blueprint for the establishment of more personalized organ chip and 'body-on-a-chip' models in the future.

9.
Cell Stem Cell ; 28(4): 587-589, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33798416

RESUMO

Methods for deriving the ureteric epithelium (UE) in vitro could improve understanding of kidney development and patterning. In this issue of Cell Stem Cell, Howden et al. (2021) identified transcriptionally distinct cell populations in human induced pluripotent stem cell (iPSC)-derived distal nephron (DN) epithelia that were inducible to UE phenotype within kidney organoids.


Assuntos
Células-Tronco Pluripotentes Induzidas , Células-Tronco Pluripotentes , Diferenciação Celular , Epitélio , Humanos , Rim , Néfrons , Organoides
10.
ACS Nano ; 13(8): 9397-9407, 2019 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-31381848

RESUMO

Graphene quantum dots (GQDs) are the harbingers of a paradigm shift that revitalize self-assembly of the colloidal puzzle by adding shape and size to the material-design palette. Although self-assembly is ubiquitous in nature, the extent to which these molecular legos can be engineered reminds us that we are still apprenticing polymer carpenters. In this quest to unlock exotic nanostructures ascending from eventual anisotropy, we have utilized different concentrations of GQDs as a filler in free-radical-mediated aqueous copolymerization. Extensive polymer grafting over the geometrically confined landscape of GQDs (0.05%) bolsters crystallization instilling a loom which steers interaction of polymeric cilia into interlaced equilateral triangles with high sophistication. Such two-dimensional (2D) assemblies epitomizing the planar tiling of "Star of David" forming a molecular kagome lattice (KL) without metal templation evoke petrichor. Interestingly, a higher percentage (0.3%) of GQDs allow selective tuning of the interfacial property of copolymers breaking symmetry due to surface energy incongruity, producing exotic Janus nanomicelles (JNMs). Herein, with the help of a suite of characterizations, we delineate the mechanism behind the formation of the KL and JNMs which forms a depot of heightened drug accretion with targeted delivery of 5-fluorouracil in the colon as validated by gamma scintigraphy studies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA