Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Synth Biol ; 13(3): 825-836, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38377949

RESUMEN

Enhancement of glucose-stimulated insulin secretion (GSIS) in exogenously delivered pancreatic ß-cells is desirable, for example, to overcome the insulin resistance manifested in type 2 diabetes or to reduce the number of ß-cells for supporting homeostasis of blood sugar in type 1 diabetes. Optogenetically engineered cells can potentiate their function with exposure to light. Given that cyclic adenosine monophosphate (cAMP) mediates GSIS, we surmised that optoamplification of GSIS is feasible in human ß-cells carrying a photoactivatable adenylyl cyclase (PAC). To this end, human EndoC-ßH3 cells were engineered to express a blue-light-activated PAC, and a workflow was established combining the scalable manufacturing of pseudoislets (PIs) with efficient adenoviral transduction, resulting in over 80% of cells carrying PAC. Changes in intracellular cAMP and GSIS were determined with the photoactivation of PAC in vitro as well as after encapsulation and implantation in mice with streptozotocin-induced diabetes. cAMP rapidly rose in ß-cells expressing PAC with illumination and quickly declined upon its termination. Light-induced amplification in cAMP was concomitant with a greater than 2-fold GSIS vs ß-cells without PAC in elevated glucose. The enhanced GSIS retained its biphasic pattern, and the rate of oxygen consumption remained unchanged. Diabetic mice receiving the engineered ß-cell PIs exhibited improved glucose tolerance upon illumination compared to those kept in the dark or not receiving cells. The findings support the use of optogenetics for molecular customization of the ß-cells toward better treatments for diabetes without the adverse effects of pharmacological approaches.


Asunto(s)
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Humanos , Ratones , Animales , Insulina , Línea Celular , Glucosa/farmacología , AMP Cíclico , Adenilil Ciclasas/genética
2.
Front Endocrinol (Lausanne) ; 14: 1212749, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37645413

RESUMEN

Introduction: Blood sugar homeostasis relies largely on the action of pancreatic islet hormones, particularly insulin and glucagon. In a prototypical fashion, glucagon is released upon hypoglycemia to elevate glucose by acting on the liver while elevated glucose induces the secretion of insulin which leads to sugar uptake by peripheral tissues. This simplified view of glucagon and insulin does not consider the paracrine roles of the two hormones modulating the response to glucose of α- and ß-cells. In particular, glucose-stimulated glucagon secretion by isolated α-cells exhibits a Hill-function pattern, while experiments with intact pancreatic islets suggest a 'U'-shaped response. Methods: To this end, a framework was developed based on first principles and coupled to experimental studies capturing the glucose-induced response of pancreatic α- and ß-cells influenced by the two hormones. The model predicts both the transient and steady-state profiles of secreted insulin and glucagon, including the typical biphasic response of normal ß-cells to hyperglycemia. Results and discussion: The results underscore insulin activity as a differentiating factor of the glucagon secretion from whole islets vs. isolated α-cells, and highlight the importance of experimental conditions in interpreting the behavior of islet cells in vitro. The model also reproduces the hyperglucagonemia, which is experienced by diabetes patients, and it is linked to a failure of insulin to inhibit α-cell activity. The framework described here is amenable to the inclusion of additional islet cell types and extrapancreatic tissue cells simulating multi-organ systems. The study expands our understanding of the interplay of insulin and glucagon for pancreas function in normal and pathological conditions.


Asunto(s)
Células Secretoras de Glucagón , Insulina , Humanos , Glucagón , Glucosa/farmacología , Hormonas Pancreáticas
3.
bioRxiv ; 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38168441

RESUMEN

We report an integrated optogenetic and bioelectronic platform for stable and long-term modulation and monitoring of cardiomyocyte function in vitro. Optogenetic inputs were achieved through expression of a photoactivatable adenylyl cyclase (bPAC), that when activated by blue light caused a dose-dependent and time-limited increase in autonomous cardiomyocyte beat rate. Bioelectronic readouts were achieved through an integrated planar multi-electrode array (MEA) that provided real-time readouts of electrophysiological activity from 32 spatially-distinct locations. Irradiation at 27 µW/mm2 resulted in a ca. 14% increase in beat rate within 20-25 minutes, which remained stable for at least 2 hours. The beating rate could be cycled through repeated "on" and "off' states, and its magnitude was a monotonic function of irradiation intensity. Our integrated platform opens new avenues in bioelectronic medicine, including closed-loop feedback systems, with potential applications for cardiac regulation including arrhythmia diagnosis and intervention.

4.
Mol Metab ; 64: 101552, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35863638

RESUMEN

BACKGROUND: Optogenetic modalities as well as optochemical and photopharmacological strategies, collectively termed optical methods, have revolutionized the control of cellular functions via light with great spatiotemporal precision. In comparison to the major advances in the photomodulation of signaling activities noted in neuroscience, similar applications to endocrine cells of the pancreas, particularly insulin-producing ß-cells, have been limited. The availability of tools allowing light-mediated changes in the trafficking of ions such as K+ and Ca2+ and signaling intermediates such as cyclic adenosine monophosphate (cAMP), renders ß-cells and their glucose-stimulated insulin secretion (GSIS) amenable to optoengineering for drug-free control of blood sugar. SCOPE OF REVIEW: The molecular circuit of the GSIS in ß-cells is described with emphasis on intermediates which are targetable for optical intervention. Various pharmacological agents modifying the release of insulin are reviewed along with their documented side effects. These are contrasted with optical approaches, which have already been employed for engineering ß-cell function or are considered for future such applications. Principal obstacles are also discussed as the implementation of optogenetics is pondered for tissue engineering and biology applications of the pancreas. MAJOR CONCLUSIONS: Notable advances in optogenetic, optochemical and photopharmacological tools are rendering feasible the smart engineering of pancreatic cells and tissues with light-regulated function paving the way for novel solutions for addressing pancreatic pathologies including diabetes.


Asunto(s)
Células Secretoras de Insulina , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Páncreas/metabolismo
5.
Cells ; 10(10)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34685602

RESUMEN

Efforts to direct the specification of human pluripotent stem cells (hPSCs) to therapeutically important somatic cell types have focused on identifying proper combinations of soluble cues. Yet, whether exosomes, which mediate intercellular communication, play a role in the differentiation remains unexplored. We took a first step toward addressing this question by subjecting hPSCs to stage-wise specification toward cardiomyocytes (CMs) in scalable stirred-suspension cultures and collecting exosomes. Samples underwent liquid chromatography (LC)/mass spectrometry (MS) and subsequent proteomic analysis revealed over 300 unique proteins from four differentiation stages including proteins such as PPP2CA, AFM, MYH9, MYH10, TRA2B, CTNNA1, EHD1, ACTC1, LDHB, and GPC4, which are linked to cardiogenic commitment. There was a significant correlation of the protein composition of exosomes with the hPSC line and stage of commitment. Differentiating hPSCs treated with exosomes from hPSC-derived CMs displayed improved efficiency of CM formation compared to cells without exogenously added vesicles. Collectively, these results demonstrate that exosomes from hPSCs induced along the CM lineage contain proteins linked to the specification process with modulating effects and open avenues for enhancing the biomanufacturing of stem cell products for cardiac diseases.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Exosomas/metabolismo , Miocitos Cardíacos , Células Madre Pluripotentes , Proteoma/metabolismo , Línea Celular , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
6.
Biotechnol Bioeng ; 118(2): 979-991, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33205831

RESUMEN

Scalable processes are requisite for the robust biomanufacturing of human pluripotent stem cell (hPSC)-derived therapeutics. Toward this end, we demonstrate the xeno-free expansion and directed differentiation of human embryonic and induced pluripotent stem cells to definitive endoderm (DE) in a controlled stirred suspension bioreactor (SSB). Based on previous work on converting hPSCs to insulin-producing progeny, differentiation of two hPSC lines was optimized in planar cultures yielding up to 87% FOXA2+ /SOX17+ cells. Next, hPSCs were propagated in an SSB with controlled pH and dissolved oxygen. Cultures displayed a 10- to 12-fold increase in cell number over 5-6 days with the maintenance of pluripotency (>85% OCT4+ ) and viability (>85%). For differentiation, SSB cultures yielded up to 89% FOXA2+ /SOX17+ cells or ~ 8 DE cells per seeded hPSC. Specification to DE cell fate was consistently more efficient in the bioreactor compared to planar cultures. Hence, a tunable strategy is established that is suitable for the xeno-free manufacturing of DE cells from different hPSC lines in scalable SSBs. This study advances bioprocess development for producing a wide gamut of human DE cell-derived therapeutics.


Asunto(s)
Reactores Biológicos , Endodermo/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Línea Celular , Endodermo/citología , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología
7.
Front Bioeng Biotechnol ; 8: 571425, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33195131

RESUMEN

Functional heart cells and tissues sourced from human pluripotent stem cells (hPSCs) have great potential for substantially advancing treatments of cardiovascular maladies. Realization of this potential will require the development of cost-effective and tunable bioprocesses for manufacturing hPSC-based cell therapeutics. Here, we report the development of a xeno-free platform for guiding the cardiogenic commitment of hPSCs. The system is based on a fully defined, open-source formulation without complex supplements, which have varied and often undetermined effects on stem cell physiology. The formulation was used to systematically investigate factors inducing the efficient commitment to cardiac mesoderm of three hPSC lines. Contractile clusters of cells appeared within a week of differentiation in planar cultures and by day 13 over 80% of the cells expressed cardiac progeny markers such as TNNT2. In conjunction with expansion, this differentiation strategy was employed in stirred-suspension cultures of hPSCs. Scalable differentiation resulted in 0.4-2 million CMs/ml or ∼5-20 TNNT2-positive cells per seeded hPSC without further enrichment. Our findings will contribute to the engineering of bioprocesses advancing the manufacturing of stem cell-based therapeutics for heart diseases.

8.
Nat Rev Endocrinol ; 16(9): 506-518, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32587391

RESUMEN

Diabetes mellitus, which affects more than 463 million people globally, is caused by the autoimmune ablation or functional loss of insulin-producing ß-cells, and prevalence is projected to continue rising over the next decades. Generating ß-cells to mitigate the aberrant glucose homeostasis manifested in the disease has remained elusive. Substantial advances have been made in producing mature ß-cells from human pluripotent stem cells that respond appropriately to dynamic changes in glucose concentrations in vitro and rapidly function in vivo following transplantation in mice. Other potential avenues to produce functional ß-cells include: transdifferentiation of closely related cell types (for example, other pancreatic islet cells such as α-cells, or other cells derived from endoderm); the engineering of non-ß-cells that are capable of modulating blood sugar; and the construction of synthetic 'cells' or particles mimicking functional aspects of ß-cells. This Review focuses on the current status of generating ß-cells via these diverse routes, highlighting the unique advantages and challenges of each approach. Given the remarkable progress in this field, scalable bioengineering processes are also discussed for the realization of the therapeutic potential of derived ß-cells.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/terapia , Células Secretoras de Insulina/fisiología , Células Madre Pluripotentes/fisiología , Células Madre/fisiología , Animales , Reactores Biológicos , Blastocisto/citología , Células Madre Embrionarias/fisiología , Humanos , Inmunosupresores , Lactante , Recién Nacido , Islotes Pancreáticos/fisiología , Ratones , Trasplante de Células Madre
9.
Front Cell Dev Biol ; 7: 235, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31696115

RESUMEN

Regenerating islet-derived (Reg) proteins have emerged as multifunctional agents with pro-proliferative, anti-apoptotic, differentiation-inducing and bactericidal properties. Over the last 40 years since first discovered, Reg proteins have been implicated in a gamut of maladies including diabetes, various types of cancer of the digestive tract, and Alzheimer disease. Surprisingly though, a consensus is still absent on the regulation of their expression, and molecular underpinning of their function. Here, we provide a critical appraisal of recent findings in the field of Reg protein biology. Specifically, the structural characteristics are reviewed particularly in connection with established or purported functions of different members of the Reg family. Moreover, Reg expression patterns in different tissues both under normal and pathophysiological conditions are summarized. Putative receptors and cascades reported to relay Reg signaling inciting cellular responses are presented aiming at a better appreciation of the biological activities of the distinct Reg moieties. Challenges are also discussed that have hampered thus far the rapid progress in this field such as the use of non-standard nomenclature for Reg molecules among various research groups, the existence of multiple Reg members with significant degree of homology and possibly compensatory modes of action, and the need for common assays with robust readouts of Reg activity. Coordinated research is warranted going forward, given that several research groups have independently linked Reg proteins to diseased states and raised the possibility that these biomolecules can serve as therapeutic targets and biomarkers.

10.
ACS Synth Biol ; 8(10): 2248-2255, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31518106

RESUMEN

Pharmacological augmentation of glucose-stimulated insulin secretion (GSIS), for example, to overcome insulin resistance in type 2 diabetes is linked to suboptimal regulation of blood sugar. Cultured ß-cells and islets expressing a photoactivatable adenylyl cyclase (PAC) are amenable to GSIS potentiation with light. However, whether PAC-mediated enhancement of GSIS can improve the diabetic state remains unknown. To this end, ß-cells were engineered with stable PAC expression that led to over 2-fold greater GSIS upon exposure to blue light while there were no changes in the absence of glucose. Moreover, the rate of oxygen consumption was unaltered despite the photoinduced elevation of GSIS. Transplantation of these cells into streptozotocin-treated mice resulted in improved glucose tolerance, lower hyperglycemia, and higher plasma insulin when subjected to illumination. Embedding optogenetic networks in ß-cells for physiologically relevant control of GSIS will enable novel solutions potentially overcoming the shortcomings of current treatments for diabetes.


Asunto(s)
AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Línea Celular , Modelos Animales de Enfermedad , Glucosa/metabolismo , Células HEK293 , Humanos , Insulina/metabolismo , Secreción de Insulina/fisiología , Luz , Ratones , Ratones Endogámicos C57BL , Optogenética/métodos , Consumo de Oxígeno/fisiología
11.
Am J Physiol Cell Physiol ; 317(2): C366-C374, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31166710

RESUMEN

Regenerating islet-derived (Reg) proteins, which were first discovered in the pancreas, are associated with increased proliferation, prevention of apoptosis, and enhanced differentiation in normal and disease states, but very little is known about the regulation of their expression. We hypothesized that Reg expression is influenced by microRNAs. Bioinformatic analysis predicted Reg1 to be a target of microRNA-7 (miR-7), which influences pancreatic ß-cell function. To this end, we investigated the effects of miR-7 on Reg1 expression in pancreatic acinar and islet ß-cells. High levels of Reg1 were noted by immunostaining and Western blotting in acinar cells in contrast to islet cells. A reciprocal expression pattern was observed for miR-7. Overexpression of miR-7 resulted in Reg1 mRNA suppression and reduction of secreted Reg1 protein. Conversely, miR-7 knockdown led to increases in Reg1. Targeting of Reg1 by miR-7 was confirmed via luciferase activity assays. In contrast, miR-7 did not directly repress the human ortholog of Reg1 REG1A as well as REG1B indicating species differences in the regulation of Reg expression. This is the first account of microRNA modulation of any Reg member warranting studies to fill gaps in our knowledge of Reg protein biology, particularly in disease contexts.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Litostatina/metabolismo , MicroARNs/metabolismo , Páncreas Exocrino/metabolismo , Regiones no Traducidas 3' , Animales , Sitios de Unión , Línea Celular Tumoral , Regulación de la Expresión Génica , Células HEK293 , Humanos , Litostatina/genética , Ratones Endogámicos C57BL , MicroARNs/genética , Páncreas Exocrino/citología , Especificidad de la Especie
12.
Curr Diab Rep ; 18(12): 133, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30343423

RESUMEN

PURPOSE OF REVIEW: Ever since the reprogramming of human fibroblasts to induced pluripotent stem cells (hiPSCs), scientists have been trying to determine if hiPSCs can give rise to progeny akin to native terminally differentiated cells as human embryonic stem cells (hESCs) do. Many different somatic cell types have been successfully reprogrammed via a variety of methods. In this review, we will discuss recent studies comparing hiPSCs and hESCs and their ability to differentiate to desired cell types as well as explore diabetes disease models. RECENT FINDINGS: Both somatic cell origin and the reprogramming method are important to the epigenetic state of the hiPSCs; however, genetic background contributes the most to differences seen between hiPSCs and hESCs. Based on our review of the relevant literature, hiPSCs display differences compared to hESCs, including a higher propensity for specification toward particular cell types based on memory retained from the somatic cell of origin. Moreover, hiPSCs provide a unique opportunity for creating diabetes disease models.


Asunto(s)
Diabetes Mellitus/patología , Diabetes Mellitus/terapia , Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Células Secretoras de Insulina/patología , Modelos Biológicos , Diferenciación Celular , Humanos
13.
ACS Biomater Sci Eng ; 3(8): 1510-1518, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28989958

RESUMEN

The development of platforms for the expansion and directed differentiation of human pluripotent stem cells (hPSCs) in large quantities under xeno-free conditions is a key step toward the realization of envisioned stem cell-based therapies. Microcarrier bioreactors afford great surface-to-volume ratio, scalability and customization with typical densities of 106-107 cells/ml or higher. In this study, a simple and inexpensive method was established for generating microcarriers without animal-derived components. While coating polystyrene beads with vitronectin alone did not support the culture of hPSCs in stirred suspension, the inclusion of recombinant human serum albumin and UV irradiation led to enhanced seeding efficiency and retention while cells grew more than 20-fold per passage for multiple successive passages and without loss of cell pluripotency. Human PSCs expanded on microcarriers were coaxed to tri-lineage differentiation demonstrating that this system can be used for the self-renewal and specification of hPSCs to therapeutically relevant cell types. Such systems will be critical for the envisioned use of stem cells in regenerative medicine and drug discovery.

14.
Sci Rep ; 7(1): 9357, 2017 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-28839233

RESUMEN

Pancreatic ß-cell insulin production is orchestrated by a complex circuitry involving intracellular elements including cyclic AMP (cAMP). Tackling aberrations in glucose-stimulated insulin release such as in diabetes with pharmacological agents, which boost the secretory capacity of ß-cells, is linked to adverse side effects. We hypothesized that a photoactivatable adenylyl cyclase (PAC) can be employed to modulate cAMP in ß-cells with light thereby enhancing insulin secretion. To that end, the PAC gene from Beggiatoa (bPAC) was delivered to ß-cells. A cAMP increase was noted within 5 minutes of photostimulation and a significant drop at 12 minutes post-illumination. The concomitant augmented insulin secretion was comparable to that from ß-cells treated with secretagogues. Greater insulin release was also observed over repeated cycles of photoinduction without adverse effects on viability and proliferation. Furthermore, the expression and activation of bPAC increased cAMP and insulin secretion in murine islets and in ß-cell pseudoislets, which displayed a more pronounced light-triggered hormone secretion compared to that of ß-cell monolayers. Calcium channel blocking curtailed the enhanced insulin response due to bPAC activity. This optogenetic system with modulation of cAMP and insulin release can be employed for the study of ß-cell function and for enabling new therapeutic modalities for diabetes.


Asunto(s)
Secreción de Insulina/efectos de la radiación , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/efectos de la radiación , Luz , Optogenética , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Animales , Línea Celular , AMP Cíclico/metabolismo , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/efectos de la radiación , Ratones , Optogenética/métodos
15.
J Biol Eng ; 11: 21, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28680477

RESUMEN

Recent advances in the expansion and directed pancreatogenic differentiation of human pluripotent stem cells (hPSCs) have intensified efforts to generate functional pancreatic islet cells, especially insulin-secreting ß-cells, for cell therapies against diabetes. However, the consistent generation of glucose-responsive insulin-releasing cells remains challenging. In this article, we first present basic concepts of pancreatic organogenesis, which frequently serves as a basis for engineering differentiation regimens. Next, past and current efforts are critically discussed for the conversion of hPSCs along pancreatic cell lineages, including endocrine ß-cells and α-cells, as well as exocrine cells with emphasis placed on the later stages of commitment. Finally, major challenges and future directions are examined, such as the identification of factors for in vivo maturation, large-scale culture and post processing systems, cell loss during differentiation, culture economics, efficiency, and efficacy and exosomes and miRNAs in pancreatic differentiation.

16.
Methods Mol Biol ; 1502: 35-52, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26659793

RESUMEN

Pluripotent stem cells can differentiate to any cell type and contribute to damaged tissue repair and organ function reconstitution. The scalable culture of pluripotent stem cells is essential to furthering the use of stem cell products in a wide gamut of applications such as screening of candidate drugs and cell replacement therapies. Human stem cell cultivation in stirred-suspension vessels enables the expansion of stem cells and the generation of differentiated progeny in quantities suitable for use in animal models and clinical studies. We describe methods of culturing human pluripotent stem cells in spinner flasks either as aggregates or on microcarriers. Techniques for assessing the quality of the culture and characterizing the cells based on the presentation of pertinent markers are also presented. Spinner flask culture with its relatively low capital and operating costs is appealing to laboratories interested in scaling up their production of stem/progenitor cells.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/citología , Agregación Celular , Técnicas de Cultivo de Célula/instrumentación , Diferenciación Celular , Línea Celular , Endodermo/citología , Endodermo/metabolismo , Citometría de Flujo/métodos , Humanos , Inmunohistoquímica/métodos , Cariotipificación/métodos , Mesodermo/citología , Mesodermo/metabolismo , Células Madre Pluripotentes/metabolismo , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos
17.
J Biotechnol ; 208: 70-9, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26036699

RESUMEN

The cultivation of stem cells as aggregates in scalable bioreactor cultures is an appealing modality for the large-scale manufacturing of stem cell products. Aggregation phenomena are central to such bioprocesses affecting the viability, proliferation and differentiation trajectory of stem cells but a quantitative framework is currently lacking. A population balance equation (PBE) model was used to describe the temporal evolution of the embryonic stem cell (ESC) cluster size distribution by considering collision-induced aggregation and cell proliferation in a stirred-suspension vessel. For ESC cultures at different agitation rates, the aggregation kernel representing the aggregation dynamics was successfully recovered as a solution of the inverse problem. The rate of change of the average aggregate size was greater at the intermediate rate tested suggesting a trade-off between increased collisions and agitation-induced shear. Results from forward simulation with obtained aggregation kernels were in agreement with transient aggregate size data from experiments. We conclude that the framework presented here can complement mechanistic studies offering insights into relevant stem cell clustering processes. More importantly from a process development standpoint, this strategy can be employed in the design and control of bioreactors for the generation of stem cell derivatives for drug screening, tissue engineering and regenerative medicine.


Asunto(s)
Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Modelos Biológicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Animales , Agregación Celular , Ratones
18.
PLoS One ; 10(4): e0123380, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25923916

RESUMEN

Genetic studies have placed the Fgfr1 gene at the top of major ontogenic pathways that enable gastrulation, tissue development and organogenesis. Using genome-wide sequencing and loss and gain of function experiments the present investigation reveals a mechanism that underlies global and direct gene regulation by the nuclear form of FGFR1, ensuring that pluripotent Embryonic Stem Cells differentiate into Neuronal Cells in response to Retinoic Acid. Nuclear FGFR1, both alone and with its partner nuclear receptors RXR and Nur77, targets thousands of active genes and controls the expression of pluripotency, homeobox, neuronal and mesodermal genes. Nuclear FGFR1 targets genes in developmental pathways represented by Wnt/ß-catenin, CREB, BMP, the cell cycle and cancer-related TP53 pathway, neuroectodermal and mesodermal programing networks, axonal growth and synaptic plasticity pathways. Nuclear FGFR1 targets the consensus sequences of transcription factors known to engage CREB-binding protein, a common coregulator of transcription and established binding partner of nuclear FGFR1. This investigation reveals the role of nuclear FGFR1 as a global genomic programmer of cell, neural and muscle development.


Asunto(s)
Núcleo Celular/metabolismo , Genoma , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Familia de Multigenes , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Análisis de Secuencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tretinoina/farmacología
19.
Tissue Eng Part B Rev ; 21(4): 377-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25813860

RESUMEN

Strategies for harnessing stem cells as a source to treat cell loss in heart disease are the subject of intense research. Human pluripotent stem cells (hPSCs) can be expanded extensively in vitro and therefore can potentially provide sufficient quantities of patient-specific differentiated cardiomyocytes. Although multiple stimuli direct heart development, the differentiation process is driven in large part by signaling activity. The engineering of hPSCs to heart cell progeny has extensively relied on establishing proper combinations of soluble signals, which target genetic programs thereby inducing cardiomyocyte specification. Pertinent differentiation strategies have relied as a template on the development of embryonic heart in multiple model organisms. Here, information on the regulation of cardiomyocyte development from in vivo genetic and embryological studies is critically reviewed. A fresh interpretation is provided of in vivo and in vitro data on signaling pathways and gene regulatory networks (GRNs) underlying cardiopoiesis. The state-of-the-art understanding of signaling pathways and GRNs presented here can inform the design and optimization of methods for the engineering of tissues for heart therapies.


Asunto(s)
Diferenciación Celular/genética , Redes Reguladoras de Genes , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Transducción de Señal/genética , Animales , Corazón/embriología , Humanos , Células Madre Pluripotentes/citología
20.
Stem Cells Dev ; 24(7): 892-903, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25405279

RESUMEN

Human pluripotent stem cells (hPSCs) display a very short G1 phase and rapid proliferation kinetics. Regulation of the cell cycle, which is linked to pluripotency and differentiation, is dependent on the stem cell environment, particularly on culture density. This link has been so far empirical and central to disparities in the growth rates and fractions of self-renewing hPSCs residing in different cycle phases. In this study, hPSC cycle progression in conjunction with proliferation and differentiation were comprehensively investigated for different culture densities. Cell proliferation decelerated significantly at densities beyond 50×10(4) cells/cm(2). Correspondingly, the G1 fraction increased from 25% up to 60% at densities greater than 40×10(4) cells/cm(2) while still hPSC pluripotency marker expression was maintained. In parallel, expression of the cycle inhibitor CDKN1A (p21) was increased, while that of p27 and p53 did not change significantly. After 4 days of culture in an unconditioned medium, greater heterogeneity was noted in the differentiation outcomes and was limited by reducing the density variation. A quantitative model was constructed for self-renewing and differentiating hPSC ensembles to gain a better understanding of the link between culture density, cycle progression, and stem cell state. Results for multiple hPSC lines and medium types corroborated experimental findings. Media commonly used for maintenance of self-renewing hPSCs exhibited the slowest kinetics of induction of differentiation (kdiff), while BMP4 supplementation led to 14-fold higher kdiff values. Spontaneous differentiation in a growth factor-free medium exhibited the largest variation in outcomes at different densities. In conjunction with the quantitative framework, our findings will facilitate rationalizing the selection of cultivation conditions for the generation of stem cell therapeutics.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Fase G1 , Células Madre Pluripotentes Inducidas/citología , Línea Celular , Medios de Cultivo Condicionados/química , Medios de Cultivo Condicionados/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Péptidos y Proteínas de Señalización Intercelular/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA