Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38334719

RESUMO

Protein therapeutics are an expanding area for research and drug development, and lipid nanoparticles (LNPs) are the most prominent nonviral vehicles for protein delivery. The most common methods for assessing protein delivery by LNPs include assays that measure the total amount of protein taken up by cells and assays that measure the phenotypic changes associated with protein delivery. However, assays for total cellular uptake include large amounts of protein that are trapped in endosomes or are otherwise nonfunctional. Assays for functional delivery are important, but the readouts are indirect and amplified, limiting the quantitative interpretation. Here, we apply an assay for cytosolic delivery, the chloroalkane penetration assay (CAPA), to measure the cytosolic delivery of a (-30) green fluorescent protein (GFP) fused to Cre recombinase (Cre(-30)GFP) fusion protein by LNPs. We compare these data to the data from total cellular uptake and functional delivery assays to provide a richer analysis of uptake and endosomal escape for LNP-mediated protein delivery. We also use CAPA for a screen of a small library of lipidoids, identifying those with a promising ability to deliver Cre(-30)GFP to the cytosol of mammalian cells. With careful controls and optimized conditions, we expect that CAPA will be a useful tool for investigating the rate, efficiency, and mechanisms of LNP-mediated delivery of therapeutic proteins.

2.
Proc Natl Acad Sci U S A ; 121(2): e2308415120, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38150477

RESUMO

Genomic DNA of the cyanophage S-2L virus is composed of 2-aminoadenine (Z), thymine (T), guanine (G), and cytosine (C), forming the genetic alphabet ZTGC, which violates Watson-Crick base pairing rules. The Z-base has an extra amino group on the two position that allows the formation of a third hydrogen bond with thymine in DNA strands. Here, we explored and expanded applications of this non-Watson-Crick base pairing in protein expression and gene editing. Both ZTGC-DNA (Z-DNA) and ZUGC-RNA (Z-RNA) produced in vitro show detectable compatibility and can be decoded in mammalian cells, including Homo sapiens cells. Z-crRNA can guide CRISPR-effectors SpCas9 and LbCas12a to cleave specific DNA through non-Watson-Crick base pairing and boost cleavage activities compared to A-crRNA. Z-crRNA can also allow for efficient gene and base editing in human cells. Together, our results help pave the way for potential strategies for optimizing DNA or RNA payloads for gene editing therapeutics and give insights to understanding the natural Z-DNA genome.


Assuntos
Pareamento de Bases , Sistemas CRISPR-Cas , DNA Forma Z , Edição de Genes , Humanos , DNA/genética , DNA/química , DNA Forma Z/genética , Edição de Genes/métodos , RNA/genética , RNA Guia de Sistemas CRISPR-Cas , Timina/química
3.
Proc Natl Acad Sci U S A ; 120(52): e2311752120, 2023 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-38134199

RESUMO

The emergence of highly transmissible severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) that are resistant to the current COVID-19 vaccines highlights the need for continued development of broadly protective vaccines for the future. Here, we developed two messenger RNA (mRNA)-lipid nanoparticle (LNP) vaccines, TU88mCSA and ALCmCSA, using the ancestral SARS-CoV-2 spike sequence, optimized 5' and 3' untranslated regions (UTRs), and LNP combinations. Our data showed that these nanocomplexes effectively activate CD4+ and CD8+ T cell responses and humoral immune response and provide complete protection against WA1/2020, Omicron BA.1 and BQ.1 infection in hamsters. Critically, in Omicron BQ.1 challenge hamster models, TU88mCSA and ALCmCSA not only induced robust control of virus load in the lungs but also enhanced protective efficacy in the upper respiratory airways. Antigen-specific immune analysis in mice revealed that the observed cross-protection is associated with superior UTRs [Carboxylesterase 1d (Ces1d)/adaptor protein-3ß (AP3B1)] and LNP formulations that elicit robust lung tissue-resident memory T cells. Strong protective effects of TU88mCSA or ALCmCSA against both WA1/2020 and VOCs suggest that this mRNA-LNP combination can be a broadly protective vaccine platform in which mRNA cargo uses the ancestral antigen sequence regardless of the antigenic drift. This approach could be rapidly adapted for clinical use and timely deployment of vaccines against emerging and reemerging VOCs.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Cricetinae , Animais , Humanos , Camundongos , RNA Mensageiro/genética , Vacinas contra COVID-19/genética , Vacinas de mRNA , SARS-CoV-2/genética , COVID-19/prevenção & controle , Regiões 3' não Traduzidas , Anticorpos Neutralizantes , Anticorpos Antivirais
4.
Nat Commun ; 14(1): 6645, 2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37863882

RESUMO

Endo-lysosomal escape is a highly inefficient process, which is a bottleneck for intracellular delivery of biologics, including proteins and nucleic acids. Herein, we demonstrate the design of a lipid-based nanoscale molecular machine, which achieves efficient cytosolic transport of biologics by destabilizing endo-lysosomal compartments through nanomechanical action upon light irradiation. We fabricate lipid-based nanoscale molecular machines, which are designed to perform mechanical movement by consuming photons, by co-assembling azobenzene lipidoids with helper lipids. We show that lipid-based nanoscale molecular machines adhere onto the endo-lysosomal membrane after entering cells. We demonstrate that continuous rotation-inversion movement of Azo lipidoids triggered by ultraviolet/visible irradiation results in the destabilization of the membranes, thereby transporting cargoes, such as mRNAs and Cre proteins, to the cytoplasm. We find that the efficiency of cytosolic transport is improved about 2.1-fold, compared to conventional intracellular delivery systems. Finally, we show that lipid-based nanoscale molecular machines are competent for cytosolic transport of tumour antigens into dendritic cells, which induce robust antitumour activity in a melanoma mouse model.


Assuntos
Produtos Biológicos , Luz , Animais , Camundongos , Transporte Biológico , Lisossomos/metabolismo , Lipídeos , Produtos Biológicos/metabolismo
5.
Cell Rep Methods ; 3(8): 100572, 2023 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-37671015

RESUMO

The ability to specifically and efficiently deliver mRNA to target locations could unlock therapeutic strategies for a range of diseases. Rhym et al.1 have developed an advanced approach for high-throughput, in vivo screening of tissue-targeting nanoparticle formulations, utilizing peptide barcoding and liquid chromatography with tandem mass spectrometry.


Assuntos
Nanopartículas , RNA Mensageiro , Cromatografia Líquida , Espectrometria de Massas em Tandem
6.
ACS Nano ; 17(16): 15231-15253, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37535899

RESUMO

During the COVID-19 pandemic, mRNA (mRNA) vaccines emerged as leading vaccine candidates in a record time. Nonreplicating mRNA (NRM) and self-amplifying mRNA (SAM) technologies have been developed into high-performing and clinically viable vaccines against a range of infectious agents, notably SARS-CoV-2. mRNA vaccines demonstrate efficient in vivo delivery, long-lasting stability, and nonexistent risk of infection. The stability and translational efficiency of in vitro transcription (IVT)-mRNA can be further increased by modulating its structural elements. In this review, we present a comprehensive overview of the recent advances, key applications, and future challenges in the field of mRNA-based vaccinology.


Assuntos
COVID-19 , Humanos , COVID-19/prevenção & controle , Pandemias/prevenção & controle , Vacinologia , SARS-CoV-2/genética , RNA Mensageiro/genética , Vacinas de mRNA
9.
Nat Biotechnol ; 2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37169967

RESUMO

Genetically engineered mouse models only capture a small fraction of the genetic lesions that drive human cancer. Current CRISPR-Cas9 models can expand this fraction but are limited by their reliance on error-prone DNA repair. Here we develop a system for in vivo prime editing by encoding a Cre-inducible prime editor in the mouse germline. This model allows rapid, precise engineering of a wide range of mutations in cell lines and organoids derived from primary tissues, including a clinically relevant Kras mutation associated with drug resistance and Trp53 hotspot mutations commonly observed in pancreatic cancer. With this system, we demonstrate somatic prime editing in vivo using lipid nanoparticles, and we model lung and pancreatic cancer through viral delivery of prime editing guide RNAs or orthotopic transplantation of prime-edited organoids. We believe that this approach will accelerate functional studies of cancer-associated mutations and complex genetic combinations that are challenging to construct with traditional models.

10.
Front Immunol ; 14: 1135815, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36969239

RESUMO

Licensed COVID-19 vaccines ameliorate viral infection by inducing production of neutralizing antibodies that bind the SARS-CoV-2 Spike protein and inhibit viral cellular entry. However, the clinical effectiveness of these vaccines is transitory as viral variants escape antibody neutralization. Effective vaccines that solely rely upon a T cell response to combat SARS-CoV-2 infection could be transformational because they can utilize highly conserved short pan-variant peptide epitopes, but a mRNA-LNP T cell vaccine has not been shown to provide effective anti-SARS-CoV-2 prophylaxis. Here we show a mRNA-LNP vaccine (MIT-T-COVID) based on highly conserved short peptide epitopes activates CD8+ and CD4+ T cell responses that attenuate morbidity and prevent mortality in HLA-A*02:01 transgenic mice infected with SARS-CoV-2 Beta (B.1.351). We found CD8+ T cells in mice immunized with MIT-T-COVID vaccine significantly increased from 1.1% to 24.0% of total pulmonary nucleated cells prior to and at 7 days post infection (dpi), respectively, indicating dynamic recruitment of circulating specific T cells into the infected lungs. Mice immunized with MIT-T-COVID had 2.8 (2 dpi) and 3.3 (7 dpi) times more lung infiltrating CD8+ T cells than unimmunized mice. Mice immunized with MIT-T-COVID had 17.4 times more lung infiltrating CD4+ T cells than unimmunized mice (7 dpi). The undetectable specific antibody response in MIT-T-COVID-immunized mice demonstrates specific T cell responses alone can effectively attenuate the pathogenesis of SARS-CoV-2 infection. Our results suggest further study is merited for pan-variant T cell vaccines, including for individuals that cannot produce neutralizing antibodies or to help mitigate Long COVID.


Assuntos
COVID-19 , SARS-CoV-2 , Camundongos , Animais , Humanos , Camundongos Transgênicos , Linfócitos T CD8-Positivos , Vacinas contra COVID-19 , COVID-19/prevenção & controle , Síndrome Pós-COVID-19 Aguda , Anticorpos Neutralizantes , Epitopos , RNA Mensageiro
11.
Trends Genet ; 39(3): 208-216, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36669950

RESUMO

There is wide interest in applying genome-editing tools to prevent, treat, and cure a variety of diseases. Since the discovery of the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) systems, these techniques have been used in combination with different delivery systems to create highly efficacious treatment options. Each delivery system has its own advantages and disadvantages and is being used for various applications. With the large number of gene-editing applications being studied but very few being brought into the clinic, we review current progress in the field, specifically where genome editing has been applied in vivo and in the clinic, and identify current challenges and areas of future growth.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Edição de Genes/métodos , Resultado do Tratamento
12.
J Am Chem Soc ; 145(1): 551-559, 2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36537880

RESUMO

Photoresponsive inhibitor and noninhibitor systems have been developed to achieve on-demand enzyme activity control. However, inhibitors are only effective for a specific and narrow range of enzymes. Noninhibitor systems usually require mutation and modification of the enzymes, leading to irreversible loss of enzymatic activities. Inspired by biological membranes, we herein report a lipidoid-based artificial compartment composed of azobenzene (Azo) lipidoids and helper lipids, which can bidirectionally regulate the activity of the encapsulated enzymes by light. In this system, the reversible photoisomerization of Azo lipidoids triggered by UV/vis light creates a continuous rotation-inversion movement, thereby enhancing the permeability of the compartment membrane and allowing substrates to pass through. Moreover, the membrane can revert to its impermeable state when light is removed. Thus, enzyme activity can be switched on and off when encapsulating enzymes in the compartments. Importantly, since neither mutation nor modification is required, negligible loss of activity is observed for the encapsulated enzymes after repeated activation and inhibition. Furthermore, this approach provides a generic strategy for controlling multiple enzymes by forgoing the use of inhibitors and may broaden the applications of enzymes in biological mechanism research and precision medicine.


Assuntos
Compostos Azo , Raios Ultravioleta , Membrana Celular , Compostos Azo/farmacologia
13.
Proc Natl Acad Sci U S A ; 119(34): e2207841119, 2022 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-35969778

RESUMO

The targeted delivery of messenger RNA (mRNA) to desired organs remains a great challenge for in vivo applications of mRNA technology. For mRNA vaccines, the targeted delivery to the lymph node (LN) is predicted to reduce side effects and increase the immune response. In this study, we explored an endogenously LN-targeting lipid nanoparticle (LNP) without the modification of any active targeting ligands for developing an mRNA cancer vaccine. The LNP named 113-O12B showed increased and specific expression in the LN compared with LNP formulated with ALC-0315, a synthetic lipid used in the COVID-19 vaccine Comirnaty. The targeted delivery of mRNA to the LN increased the CD8+ T cell response to the encoded full-length ovalbumin (OVA) model antigen. As a result, the protective and therapeutic effect of the OVA-encoding mRNA vaccine on the OVA-antigen-bearing B16F10 melanoma model was also improved. Moreover, 113-O12B encapsulated with TRP-2 peptide (TRP2180-188)-encoding mRNA also exhibited excellent tumor inhibition, with the complete response of 40% in the regular B16F10 tumor model when combined with anti-programmed death-1 (PD-1) therapy, revealing broad application of 113-O12B from protein to peptide antigens. All the treated mice showed long-term immune memory, hindering the occurrence of tumor metastatic nodules in the lung in the rechallenging experiments that followed. The enhanced antitumor efficacy of the LN-targeting LNP system shows great potential as a universal platform for the next generation of mRNA vaccines.


Assuntos
Vacinas Anticâncer , Nanopartículas , Neoplasias , Vacinas de mRNA , Amino Álcoois , Animais , Antígenos/metabolismo , Linfócitos T CD8-Positivos , Vacinas Anticâncer/uso terapêutico , Decanoatos , Memória Imunológica , Lipossomos , Linfonodos , Camundongos , Metástase Neoplásica/prevenção & controle , Neoplasias/terapia , Ovalbumina , Vacinas de mRNA/uso terapêutico
14.
Langmuir ; 38(30): 9092-9098, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35852946

RESUMO

The field of nanomedicine has rapidly grown in the past decades. Although a few nanomedicines are available in the market for clinical use, it is still challenging to develop nanomedicine targeting tissues beyond the liver. It has been recognized that even though the nanoparticles are modified with targeting ligands, the formation of a protein corona on the surface of nanoparticles in the biological fluids results in limited progress in nanoparticle-based drug delivery to specific cells or tissues. In this Perspective, we will discuss the role of surface properties in determining the formation of the protein corona and summarize the recent progress in engineering the nano/bio interface for protein-corona-mediated cell- and organ-selective drug delivery. Moreover, current challenges in the field and insights into designing new strategies for targeting drug delivery with a better understanding of the protein corona will be discussed.


Assuntos
Nanopartículas , Coroa de Proteína , Sistemas de Liberação de Medicamentos , Engenharia , Nanomedicina/métodos
15.
Acta Pharm Sin B ; 12(6): 2624-2639, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35755280

RESUMO

Lipid nanoparticle (LNP)-based drug delivery systems have become the most clinically advanced non-viral delivery technology. LNPs can encapsulate and deliver a wide variety of bioactive agents, including the small molecule drugs, proteins and peptides, and nucleic acids. However, as the physicochemical properties of small- and macromolecular cargos can vary drastically, every LNP carrier system needs to be carefully tailored in order to deliver the cargo molecules in a safe and efficient manner. Our group applied the combinatorial library synthesis approach and in vitro and in vivo screening strategy for the development of LNP delivery systems for drug delivery. In this Review, we highlight our recent progress in the design, synthesis, characterization, evaluation, and optimization of combinatorial LNPs with novel structures and properties for the delivery of small- and macromolecular therapeutics both in vitro and in vivo. These delivery systems have enormous potentials for cancer therapy, antimicrobial applications, gene silencing, genome editing, and more. We also discuss the key challenges to the mechanistic study and clinical translation of new LNP-enabled therapeutics.

16.
Adv Drug Deliv Rev ; 186: 114356, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35595022

RESUMO

With the emerging advances in utilizing nanocarriers for biomedical applications, a molecular-level understanding of the in vivo fate of nanocarriers is necessary. After administration into human fluids, nanocarriers can attract proteins onto their surfaces, forming an assembled adsorption layer called protein corona (PC). The formed PC can influence the physicochemical properties and subsequently determine nanocarriers' biological behaviors. Therefore, an in-depth understanding of the features and effects of the PC on the nanocarriers' surface is the first and most important step towards controlling their in vivo fate. This review introduces fundamental knowledge such as the definition, formation, composition, conformation, and characterization of the PC, emphasizing the in vivo environmental factors that control the PC formation. The effect of PC on the physicochemical properties and thus biological behaviors of nanocarriers was then presented and thoroughly discussed. Finally, we proposed the design strategies available for engineering PC onto nanocarriers to manipulate them with the desired surface properties and achieve the best biomedical outcomes.


Assuntos
Nanopartículas , Coroa de Proteína , Humanos , Nanopartículas/química , Coroa de Proteína/química , Proteínas/química , Propriedades de Superfície
17.
Annu Rev Biomed Eng ; 24: 85-109, 2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35231177

RESUMO

mRNA vaccines have brought about a great revolution in the vaccine fields owing to their simplicity and adaptability in antigen design, potential to induce both humoral and cell-mediated immune responses and demonstrated high efficacy, and rapid and low-cost production by using the same manufacturing platform for different mRNA vaccines. Multiple mRNA vaccines have been investigated for both infectious diseases and cancers, showing significant superiority to other types of vaccines. Although great success of mRNA vaccines has been achieved in the control of the coronavirus disease 2019 pandemic, there are still multiple challenges for the future development of mRNA vaccines. In this review, the most recent developments of mRNA vaccines against both infectious diseases and cancers are summarized for an overview of this field. Moreover, the challenges are also discussed on the basis of these developments.


Assuntos
COVID-19 , Doenças Transmissíveis , Neoplasias , COVID-19/prevenção & controle , Humanos , RNA Mensageiro/genética , Vacinas Sintéticas , Vacinas de mRNA
18.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35173043

RESUMO

Safe and efficacious systemic delivery of messenger RNA (mRNA) to specific organs and cells in vivo remains the major challenge in the development of mRNA-based therapeutics. Targeting of systemically administered lipid nanoparticles (LNPs) coformulated with mRNA has largely been confined to the liver and spleen. Using a library screening approach, we identified that N-series LNPs (containing an amide bond in the tail) are capable of selectively delivering mRNA to the mouse lung, in contrast to our previous discovery that O-series LNPs (containing an ester bond in the tail) that tend to deliver mRNA to the liver. We analyzed the protein corona on the liver- and lung-targeted LNPs using liquid chromatography-mass spectrometry and identified a group of unique plasma proteins specifically absorbed onto the surface that may contribute to the targetability of these LNPs. Different pulmonary cell types can also be targeted by simply tuning the headgroup structure of N-series LNPs. Importantly, we demonstrate here the success of LNP-based RNA therapy in a preclinical model of lymphangioleiomyomatosis (LAM), a destructive lung disease caused by loss-of-function mutations in the Tsc2 gene. Our lung-targeting LNP exhibited highly efficient delivery of the mouse tuberous sclerosis complex 2 (Tsc2) mRNA for the restoration of TSC2 tumor suppressor in tumor and achieved remarkable therapeutic effect in reducing tumor burden. This research establishes mRNA LNPs as a promising therapeutic intervention for the treatment of LAM.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Linfangioleiomiomatose/tratamento farmacológico , RNA Mensageiro/administração & dosagem , Animais , Feminino , Técnicas de Transferência de Genes , Engenharia Genética/métodos , Lipossomos/química , Lipossomos/farmacologia , Pulmão/citologia , Pulmão/patologia , Pneumopatias/tratamento farmacológico , Pneumopatias/metabolismo , Linfangioleiomiomatose/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/química , Coroa de Proteína/química , Coroa de Proteína/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/farmacologia , RNA Interferente Pequeno/metabolismo
19.
ACS Biomater Sci Eng ; 8(2): 722-733, 2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35104103

RESUMO

Chimeric antigen receptor (CAR)-engineered adoptive cell therapy marks a revolution in cancer treatment based on the highly successful responses to CAR T cell therapy in the treatment of blood cancers. Due to the versatile structure of CARs, this technology can be easily adapted to other immune cell types, including macrophages and NKs, and applied in the treatment of many other cancers. However, high costs and fatal adverse effects represent significant concerns for future development. In vitro transcribed (IVT) mRNA therapeutics, which possess a high safety profile and straightforward production methods, could provide a useful alternative for CAR cell construction. However, the low stability and transfection efficiency of IVT-mRNA in immune cells limit further applications. In this work, we successfully engineered CAR macrophages (CAR-Ms) and CAR T cells with CAR mRNA using lipid nanoparticles (LNPs). Both the LNP formulations and mRNA modifications were optimized for in vitro mRNA transfection. More importantly, the CAR macrophages and CAR T cells both demonstrated significant cytotoxic effects on B lymphoma in vitro, underscoring the great potential of mRNA-engineered adoptive cell therapy.


Assuntos
Receptores de Antígenos Quiméricos , Lipossomos , Macrófagos/metabolismo , Nanopartículas , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linfócitos T/metabolismo
20.
J Control Release ; 345: 176-189, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35157939

RESUMO

Peptide- and protein-based therapeutics have drawn significant attention over the past few decades for the treatment of infectious diseases, genetic disorders, oncology, and many other clinical needs. Yet, protecting peptide- and protein-based drugs from degradation and denaturation during processing, storage and delivery remain significant challenges. In this review, we introduce the properties of peptide- and protein-based drugs and the challenges associated with their stability and delivery. Then, we discuss delivery strategies using synthetic polymers and their advantages and limitations. This is followed by a focus on silk protein-based materials for peptide/protein drug processing, storage, and delivery, as a path to overcome stability and delivery challenges with current systems.


Assuntos
Sistemas de Liberação de Medicamentos , Seda , Hidrogéis , Peptídeos , Polímeros
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...