Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 182
Filtrar
1.
Microbiol Res ; 254: 126901, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34700186

RESUMEN

In the light of intensification of cropping practices and changing climatic conditions, nourishing a growing global population requires optimizing environmental sustainability and reducing ecosystem impacts of food production. The use of microbiological systems to ameliorate the agricultural production in a sustainable and eco-friendly way is widespread accepted as a future key-technology. However, the multitude of interaction possibilities between the numerous beneficial microbes and plants in their habitat calls for systematic analysis and management of the rhizospheric microbiome. This review exploits present and future strategies for rhizospheric microbiome management with the aim to generate a comprehensive understanding of the known tools and techniques. Significant information on the structure and dynamics of rhizospheric microbiota of isolated microbial communities is now available. These microbial communities have beneficial effects including increased plant growth, essential nutrient acquisition, pathogens tolerance, and increased abiotic as well as biotic stress tolerance such as drought, temperature, salinity and antagonistic activities against the phyto-pathogens. A better and comprehensive understanding of the various effects and microbial interactions can be gained by application of molecular approaches as extraction of DNA/RNA and other biochemical markers to analyze microbial soil diversity. Novel techniques like interactome network analysis and split-ubiquitin system framework will enable to gain more insight into communication and interactions between the proteins from microbes and plants. The aim of the analysis tasks leads to the novel approach of Rhizosphere microbiome engineering. The capability of forming the rhizospheric microbiome in a defined way will allow combining several microbes (e.g. bacteria and fungi) for a given environment (soil type and climatic zone) in order to exert beneficial influences on specific plants. This integration will require a large-scale effort among academic researchers, industry researchers and farmers to understand and manage interactions of plant-microbiomes within modern farming systems, and is clearly a multi-domain approach and can be mastered only jointly by microbiology, mathematics and information technology. These innovations will open up a new avenue for designing and implementing intensive farming microbiome management approaches to maximize resource productivity and stress tolerance of agro-ecosystems, which in return will create value to the increasing worldwide population, for both food production and consumption.


Asunto(s)
Agricultura , Microbiota , Rizosfera , Desarrollo Sostenible , Agricultura/tendencias , Bioingeniería/tendencias , Productos Agrícolas/microbiología , Interacciones Microbianas , Microbiología del Suelo , Desarrollo Sostenible/tendencias
2.
Nat Rev Genet ; 23(3): 154-168, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34611352

RESUMEN

Modern genome-scale methods that identify new genes, such as proteogenomics and ribosome profiling, have revealed, to the surprise of many, that overlap in genes, open reading frames and even coding sequences is widespread and functionally integrated into prokaryotic, eukaryotic and viral genomes. In parallel, the constraints that overlapping regions place on genome sequences and their evolution can be harnessed in bioengineering to build more robust synthetic strains and constructs. With a focus on overlapping protein-coding and RNA-coding genes, this Review examines their discovery, topology and biogenesis in the context of their genome biology. We highlight exciting new uses for sequence overlap to control translation, compress synthetic genetic constructs, and protect against mutation.


Asunto(s)
Bioingeniería , Genes Sobrepuestos/fisiología , Genoma/genética , Animales , Bioingeniería/métodos , Bioingeniería/tendencias , Mapeo Cromosómico , Humanos , Organismos Modificados Genéticamente/genética
4.
Elife ; 102021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34927583

RESUMEN

Employing concepts from physics, chemistry and bioengineering, 'learning-by-building' approaches are becoming increasingly popular in the life sciences, especially with researchers who are attempting to engineer cellular life from scratch. The SynCell2020/21 conference brought together researchers from different disciplines to highlight progress in this field, including areas where synthetic cells are having socioeconomic and technological impact. Conference participants also identified the challenges involved in designing, manipulating and creating synthetic cells with hierarchical organization and function. A key conclusion is the need to build an international and interdisciplinary research community through enhanced communication, resource-sharing, and educational initiatives.


Asunto(s)
Células Artificiales , Bioingeniería/métodos , Bioingeniería/estadística & datos numéricos , Bioingeniería/tendencias , Colaboración Intersectorial , Orgánulos/fisiología , Biología Sintética/tendencias , Predicción , Humanos
5.
Cells ; 10(6)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203713

RESUMEN

Cardiovascular disease remains the leading cause of morbidity and mortality in the world. Thus, therapeutic interventions to circumvent this growing burden are of utmost importance. Extracellular vesicles (EVs) actively secreted by most living cells, play a key role in paracrine and endocrine intercellular communication via exchange of biological molecules. As the content of secreted EVs reflect the physiology and pathology of the cell of their origin, EVs play a significant role in cellular homeostasis, disease pathogenesis and diagnostics. Moreover, EVs are gaining popularity in clinics as therapeutic and drug delivery vehicles, transferring bioactive molecules such as proteins, genes, miRNAs and other therapeutic agents to target cells to treat diseases and deter disease progression. Despite our limited but growing knowledge of EV biology, it is imperative to understand the complex mechanisms of EV cargo sorting in pursuit of designing next generation EV-based therapeutic delivery systems. In this review, we highlight the mechanisms of EV cargo sorting and methods of EV bioengineering and discuss engineered EVs as a potential therapeutic delivery system to treat cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/terapia , Sistemas de Liberación de Medicamentos/métodos , Vesículas Extracelulares/fisiología , Bioingeniería/métodos , Bioingeniería/tendencias , Transporte Biológico/fisiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Comunicación Celular , Movimiento Celular , Exosomas/fisiología , Vesículas Extracelulares/metabolismo , Humanos , Transporte de Proteínas/fisiología , Proteínas/metabolismo
7.
Carbohydr Polym ; 261: 117884, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33766371

RESUMEN

Due to over-consumption of fossil resources and environmental problems, lignocellulosic biomass as the most abundant and renewable materials is considered as the best candidate to produce biomaterials, biochemicals, and bioenergy, which is of strategic significance and meets the theme of Green Chemistry. Highly efficient and green fractionation of lignocellulose components significantly boosts the high-value utilization of lignocellulose and the biorefinery development. However, heterogeneity of lignocellulosic structure severely limited the lignocellulose fractionation. This paper offers the summary and perspective of the extensive investigation that aims to give insight into the lignocellulose prior-fractionation. Based on the role and structure of lignocellulose component in the plant cell wall, lignocellulose prior-fractionation can be divided into cellulose-first strategy, hemicelluloses-first strategy, and lignin-first strategy, which realizes the selective dissociation and transformation of a component in lignocellulose. Ultimately, the challenges and opportunities of lignocellulose prior-fractionation are proposed on account of the existing problems in the biorefining valorization.


Asunto(s)
Bioquímica/tendencias , Materiales Biocompatibles/síntesis química , Bioingeniería/tendencias , Metabolismo Energético , Lignina/química , Bioquímica/métodos , Materiales Biocompatibles/química , Materiales Biocompatibles/aislamiento & purificación , Biodegradación Ambiental , Bioingeniería/métodos , Biocombustibles , Biomasa , Celulosa/química , Fraccionamiento Químico , Metabolismo Energético/fisiología , Equipo Reutilizado , Tecnología Química Verde/métodos , Tecnología Química Verde/tendencias , Humanos
8.
Semin Cancer Biol ; 74: 62-78, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33609665

RESUMEN

Cancer is a disease that evolves continuously with unpredictable outcomes. Although conventional chemotherapy can display significant antitumor effects, the lack of specificity and poor bioavailability remain major concerns in cancer therapy. Moreover, with the advent of novel anti-cancer gene therapies, there is an urgent need for drug delivery vectors capable of bypassing cellular barriers and efficiently transferring therapeutic cargo to recipient cells. A number of drug delivery systems have been proposed to overcome these limitations, but their successful clinical translation has been hampered by the onset of unexpected side effects and associated toxicities. The application of extracellular vesicles (EVs), a class of naturally released, cell-derived particles, as drug delivery vectors presents a breakthrough in nanomedicine, taking into account their biocompatibility and natural role in intercellular communication. Combining the advantageous intrinsic properties of EVs with surface functionalization and the encapsulation of drugs allows for a new class of engineered EVs that serve as effective therapeutic carriers. Here, we describe the various successful approaches involving the application of engineered EVs as bio-derived drug delivery vectors in cancer therapy. The latest and most effective strategies of engineering EVs to improve drug loading, stealth properties and tumour targeting capabilities of EVs are debated. Finally, current obstacles and future perspectives of smart engineered EVs are discussed.


Asunto(s)
Bioingeniería/métodos , Portadores de Fármacos , Sistemas de Liberación de Medicamentos/métodos , Vesículas Extracelulares , Neoplasias/tratamiento farmacológico , Animales , Bioingeniería/tendencias , Sistemas de Liberación de Medicamentos/tendencias , Humanos
9.
Biochem Soc Trans ; 49(1): 187-201, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33544117

RESUMEN

The human gut microbiome has emerged as a key player in the bidirectional communication of the gut-brain axis, affecting various aspects of homeostasis and pathophysiology. Until recently, the majority of studies that seek to explore the mechanisms underlying the microbiome-gut-brain axis cross-talk, relied almost exclusively on animal models, and particularly gnotobiotic mice. Despite the great progress made with these models, various limitations, including ethical considerations and interspecies differences that limit the translatability of data to human systems, pushed researchers to seek for alternatives. Over the past decades, the field of in vitro modelling of tissues has experienced tremendous growth, thanks to advances in 3D cell biology, materials, science and bioengineering, pushing further the borders of our ability to more faithfully emulate the in vivo situation. The discovery of stem cells has offered a new source of cells, while their use in generating gastrointestinal and brain organoids, among other tissues, has enabled the development of novel 3D tissues that better mimic the native tissue structure and function, compared with traditional assays. In parallel, organs-on-chips technology and bioengineered tissues have emerged as highly promising alternatives to animal models for a wide range of applications. Here, we discuss how recent advances and trends in this area can be applied in host-microbe and host-pathogen interaction studies. In addition, we highlight paradigm shifts in engineering more robust human microbiome-gut-brain axis models and their potential to expand our understanding of this complex system and hence explore novel, microbiome-based therapeutic approaches.


Asunto(s)
Eje Cerebro-Intestino/fisiología , Microbiota/fisiología , Técnicas de Cultivo de Tejidos/tendencias , Animales , Bioingeniería/métodos , Bioingeniería/tendencias , Células Cultivadas , Microbioma Gastrointestinal/fisiología , Humanos , Ratones , Modelos Biológicos , Organoides , Técnicas de Cultivo de Tejidos/métodos
10.
Trends Biotechnol ; 39(4): 323-327, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33573847

RESUMEN

The EU Horizon2020 consortium PHOTOFUEL joined academic and industrial partners from biology, chemistry, engineering, engine design, and lifecycle assessment, making tremendous progress towards engine-ready fuels from CO2 via engineered photosynthetic microbes. Technical, environmental, economic, and societal opportunities and challenges were explored to frame future technology realization at scale.


Asunto(s)
Bioingeniería , Biocombustibles , Luz Solar , Biocatálisis , Bioingeniería/tendencias , Fotosíntesis
11.
Nat Commun ; 12(1): 388, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33452260

RESUMEN

The practices of synthetic biology are being integrated into 'multiscale' designs enabling two-way communication across organic and inorganic information substrates in biological, digital and cyber-physical system integrations. Novel applications of 'bio-informational' engineering will arise in environmental monitoring, precision agriculture, precision medicine and next-generation biomanufacturing. Potential developments include sentinel plants for environmental monitoring and autonomous bioreactors that respond to biosensor signaling. As bio-informational understanding progresses, both natural and engineered biological systems will need to be reimagined as cyber-physical architectures. We propose that a multiple length scale taxonomy will assist in rationalizing and enabling this transformative development in engineering biology.


Asunto(s)
Bioingeniería/tendencias , Predicción , Biología Sintética/tendencias , Bioingeniería/métodos , Biología Sintética/métodos
12.
Trends Parasitol ; 37(5): 401-413, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33485788

RESUMEN

Plasmodium falciparum pathogenesis is complex and intimately connected to vascular physiology. This is exemplified by cerebral malaria (CM), a neurovascular complication that accounts for most of the malaria deaths worldwide. P. falciparum sequestration in the brain microvasculature is a hallmark of CM and is not replicated in animal models. Numerous aspects of the disease are challenging to fully understand from clinical studies, such as parasite binding tropism or causal pathways in blood-brain barrier breakdown. Recent bioengineering approaches allow for the generation of 3D microvessels and organ-specific vasculature that provide precise control of vessel architecture and flow dynamics, and hold great promise for malaria research. Here, we discuss recent and future applications of bioengineered microvessels in malaria pathogenesis research.


Asunto(s)
Bioingeniería , Microvasos , Parasitología , Plasmodium falciparum , Animales , Bioingeniería/tendencias , Encéfalo/parasitología , Humanos , Microvasos/química , Microvasos/parasitología , Parasitología/métodos , Plasmodium falciparum/fisiología
13.
Endocrinology ; 162(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33411885

RESUMEN

Extracellular vesicles (EVs) are key players of intercellular communication in the physiological and pathological setting. In cancer, EVs mediate complex signaling mechanisms between cancer cells and the tumor microenvironment (TME), and can influence tumor progression and the response to existing therapies. Importantly, EVs can be loaded with therapeutic agents and modified to display tumor-targeting molecules. In the field of nanomedicine, EVs have been engineered to serve as therapeutic delivery vehicles for several anticancer agents, including antibodies, chemotherapy, compounds, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats-associated endonuclease 9), and small interfering RNA (siRNA). Notably, the engineered EVs were shown to suppress malignant features of cancer cells, to elicit antitumor immunity, and to decrease tumor angiogenesis. Here, we review the EV-based therapies designed to target cancer cells and to educate components of the TME to drive antitumor responses. These studies illustrate the multifunctional applications of EVs in the development of anticancer therapies and their translational potential for cancer treatment.


Asunto(s)
Sistemas de Liberación de Medicamentos , Vesículas Extracelulares/fisiología , Neoplasias/terapia , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Bioingeniería/métodos , Bioingeniería/tendencias , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/fisiología , Humanos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Neoplasias/metabolismo , Neoplasias/patología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacocinética , Microambiente Tumoral/efectos de los fármacos
14.
Drug Discov Today ; 26(4): 902-915, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33383213

RESUMEN

Smart nanocarriers obtained from bacteria and viruses offer excellent biomimetic properties which has led to significant research into the creation of advanced biomimetic materials. Their versatile biomimicry has application as biosensors, biomedical scaffolds, immobilization, diagnostics, and targeted or personalized treatments. The inherent natural traits of biomimetic and bioinspired bacteria- and virus-derived nanovesicles show potential for their use in clinical vaccines and novel therapeutic drug delivery systems. The past few decades have seen significant progress in the bioengineering of bacteria and viruses to manipulate and enhance their therapeutic benefits. From a pharmaceutical perspective, biomimetics enable the safe integration of naturally occurring bacteria and virus particles to achieve high, stable rates of cellular transfection/infection and prolonged circulation times. In addition, biomimetic technologies can overcome safety concerns associated with live-attenuated and inactivated whole bacteria or viruses. In this review, we provide an update on the utilization of bacterial and viral particles as drug delivery systems, theranostic carriers, and vaccine/immunomodulation modalities.


Asunto(s)
Bioingeniería/tendencias , Materiales Biomiméticos/farmacología , Portadores de Fármacos/farmacología , Descubrimiento de Drogas/tendencias , Nanoestructuras/uso terapéutico , Fenómenos Fisiológicos Bacterianos , Biomimética , Sistemas de Liberación de Medicamentos/tendencias , Humanos , Vacunas/farmacología , Fenómenos Fisiológicos de los Virus
16.
Cells ; 9(10)2020 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-32998444

RESUMEN

The transcription factor "Kruppel-like factor 4" (KLF4) is a central player in the field of pluripotent stem cell biology. In particular, it was put under the spotlight as one of the four factors of the cocktail originally described for reprogramming into induced pluripotent stem cells (iPSCs). In contrast, its possible functions in native tissue stem cells remain largely unexplored. We recently published that KLF4 is a regulator of "stemness" in human keratinocytes. We show that reducing the level of expression of this transcription factor by RNA interference or pharmacological repression promotes the ex vivo amplification and regenerative capacity of two types of cells of interest for cutaneous cell therapy: native keratinocyte stem and progenitor cells from adult epidermis, which have been used for more than three decades in skin graft bioengineering, and keratinocytes generated by the lineage-oriented differentiation of embryonic stem cells (ESCs), which have potential for the development of skin bio-bandages. At the mechanistic level, KLF4 repression alters the expression of a large set of genes involved in TGF-ß1 and WNT signaling pathways. Major regulators of TGF-ß bioavailability and different TGF-ß receptors were targeted, notably modulating the ALK1/Smad1/5/9 axis. At a functional level, KLF4 repression produced an antagonist effect on TGFß1-induced keratinocyte differentiation.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , Piel Artificial , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta1/genética , Receptores de Activinas Tipo II/genética , Bioingeniería/tendencias , Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Factor 4 Similar a Kruppel , Interferencia de ARN , Proteína Smad1/genética , Vía de Señalización Wnt/genética
18.
Theranostics ; 10(16): 7034-7052, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32641977

RESUMEN

This review provides an update for the international research community on the cell modeling tools that could accelerate the understanding of SARS-CoV-2 infection mechanisms and could thus speed up the development of vaccines and therapeutic agents against COVID-19. Many bioengineering groups are actively developing frontier tools that are capable of providing realistic three-dimensional (3D) models for biological research, including cell culture scaffolds, microfluidic chambers for the culture of tissue equivalents and organoids, and implantable windows for intravital imaging. Here, we review the most innovative study models based on these bioengineering tools in the context of virology and vaccinology. To make it easier for scientists working on SARS-CoV-2 to identify and apply specific tools, we discuss how they could accelerate the discovery and preclinical development of antiviral drugs and vaccines, compared to conventional models.


Asunto(s)
Antivirales/aislamiento & purificación , Antivirales/farmacología , Betacoronavirus , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Vacunas Virales/aislamiento & purificación , Vacunas Virales/farmacología , Betacoronavirus/química , Betacoronavirus/genética , Betacoronavirus/inmunología , Bioingeniería/métodos , Bioingeniería/tendencias , Reactores Biológicos , COVID-19 , Vacunas contra la COVID-19 , Técnicas de Cultivo de Célula , Simulación por Computador , Infecciones por Coronavirus/inmunología , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/tendencias , Evaluación de Medicamentos/métodos , Evaluación de Medicamentos/tendencias , Farmacorresistencia Viral , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Modelos Biológicos , Organoides/citología , Organoides/virología , Neumonía Viral/inmunología , SARS-CoV-2 , Nanomedicina Teranóstica
19.
Crit Rev Immunol ; 40(1): 53-74, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32421979

RESUMEN

Therapeutic antibodies and nanotherapeutic drugs are of great concern due to their widespread use against numerous diseases worldwide. They are frequently used for targeted therapy under the assumption that they cause fewer side effects than nontargeted drugs. Despite their specificity and particular design for therapeutic actions, they might still exhibit unintended adverse effects in the immune system. Immunotoxicity reactions are mediated by immunomodulation, including immunostimulation and immunosuppression. The present review gives an overview on the adverse immunotoxic effects induced by therapeutic antibodies as well as nanotherapeutic drugs. In this context, future methods combining more efficient drug design with better tolerability and fewer adverse effects are discussed to ensure improved safety in the engineering of therapeutic antibodies and nanotherapeutics.


Asunto(s)
Anticuerpos/efectos adversos , Sistema Inmunológico/efectos de los fármacos , Nanopartículas/efectos adversos , Animales , Anticuerpos/uso terapéutico , Bioingeniería/tendencias , Citotoxicidad Inmunológica , Diseño de Fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Humanos , Inmunomodulación , Nanopartículas/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...