Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 7.518
Filtrar
1.
Clin Chim Acta ; 564: 119906, 2025 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-39127296

RESUMO

Mycoplasma pneumoniae can cause respiratory infections and pneumonia, posing a serious threat to the health of children and adolescents. Early diagnosis of Mycoplasma pneumoniae infection is crucial for clinical treatment. Currently, diagnostic methods for Mycoplasma pneumoniae infection include pathogen detection, molecular biology techniques, and bacterial culture, all of which have certain limitations. Here, we developed a rapid, simple, and accurate detection method for Mycoplasma pneumoniae that does not rely on large equipment or complex operations. This technology combines the CRISPR-Cas12a system with recombinase polymerase amplification (RPA), allowing the detection results to be observed through fluorescence curves and immunochromatographic lateral flow strips.It has been validated that RPA-CRISPR/Cas12a fluorescence analysis and RPA-CRISPR/Cas12-immunochromatographic exhibit no cross-reactivity with other common pathogens, and The established detection limit was ascertained to be as low as 102 copies/µL.Additionally, 49 clinical samples were tested and compared with fluorescence quantitative polymerase chain reaction, demonstrating a sensitivity and specificity of 100%. This platform exhibits promising clinical performance and holds significant potential for clinical application, particularly in settings with limited resources, such as clinical care points or resource-constrained areas.


Assuntos
Sistemas CRISPR-Cas , Mycoplasma pneumoniae , Mycoplasma pneumoniae/genética , Mycoplasma pneumoniae/isolamento & purificação , Humanos , Sistemas CRISPR-Cas/genética , Técnicas de Amplificação de Ácido Nucleico/métodos , Pneumonia por Mycoplasma/diagnóstico , Pneumonia por Mycoplasma/microbiologia
2.
Methods Mol Biol ; 2854: 41-50, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192117

RESUMO

The innate immune system relies on a variety of pathogen recognition receptors (PRRs) as the first line of defense against pathogenic invasions. Viruses have evolved multiple strategies to evade the host immune system through coevolution with hosts. The CRISPR-Cas system is an adaptive immune system in bacteria or archaea that defends against viral reinvasion by targeting nucleic acids for cleavage. Based on the characteristics of Cas proteins and their variants, the CRISPR-Cas system has been developed into a versatile gene-editing tool capable of gene knockout or knock-in operations to achieve genetic variations in organisms. It is now widely used in the study of viral immune evasion mechanisms. This chapter will introduce the use of the CRISPR-Cas9 system for editing herpes simplex virus 1 (HSV-1) genes to explore the mechanisms by which HSV-1 evades host innate immunity and the experimental procedures involved.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Herpesvirus Humano 1 , Evasão da Resposta Imune , Imunidade Inata , Sistemas CRISPR-Cas/genética , Imunidade Inata/genética , Herpesvirus Humano 1/imunologia , Herpesvirus Humano 1/genética , Evasão da Resposta Imune/genética , Humanos , Edição de Genes/métodos , Animais , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Herpes Simples/imunologia , Herpes Simples/virologia , Herpes Simples/genética
3.
Methods Mol Biol ; 2854: 51-60, 2025.
Artigo em Inglês | MEDLINE | ID: mdl-39192118

RESUMO

The application of CRISPR-mediated library screening has fundamentally transformed functional genomics by revealing the complexity of virus-host interactions. This protocol describes the use of CRISPR-mediated library screening to identify key functional genes regulating the innate immune response to PEDV infection. We detail a step-by-step process, starting from the design and construction of a customized CRISPR knockout library targeting genes involved in innate immunity to the effective delivery of these constructs into cells using lentiviral vectors. Subsequently, we outline the process of identifying functional genes postviral attack, including the use of next-generation sequencing (NGS), to analyze and identify knockout cells that exhibit altered responses to infection. This integrated approach provides researchers in immunology and virology with a resource and a robust framework for uncovering the genetic basis of host-pathogen interactions and the arsenal of the innate immune system against viral invasions.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes , Biblioteca Gênica , Imunidade Inata , Imunidade Inata/genética , Sistemas CRISPR-Cas/genética , Humanos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/genética , Linhagem Celular , Lentivirus/genética
4.
Carbohydr Polym ; 345: 122561, 2024 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-39227100

RESUMO

The digestibility of starch is affected by amylose content, and increasing amylopectin chain length which can be manipulated by alterations to genes encoding starch-branching enzymes (SBEs). We investigated the impact of Cas9-mediated mutagenesis of SBEs in potato on starch structural properties and digestibility. Four potato starches with edited SBE genes were tested. One lacked SBE1 and SBE2, two lacked SBE2 and had reduced SBE1, and one had reduced SBE2 only. Starch structure and thermal properties were characterised by DSC and XRD. The impact of different thermal treatments on digestibility was studied using an in vitro digestion protocol. All native potato starches were resistant to digestion, and all gelatinised starches were highly digestible. SBE modified starches had higher gelatinisation temperatures than wild type potatoes and retrograded more rapidly. Gelatinisation and 18 h of retrogradation, increased gelatinisation enthalpy, but this did not translate to differences in digestion. Following 7 days of retrogradation, starch from three modified SBE starch lines was less digestible than starch from wild-type potatoes, likely due to the recrystallisation of the long amylopectin chains. Our results indicate that reductions in SBE in potato may be beneficial to health by increasing the amount of fibre reaching the colon after retrogradation.


Assuntos
Enzima Ramificadora de 1,4-alfa-Glucana , Mutagênese , Solanum tuberosum , Amido , Solanum tuberosum/genética , Solanum tuberosum/química , Enzima Ramificadora de 1,4-alfa-Glucana/genética , Enzima Ramificadora de 1,4-alfa-Glucana/metabolismo , Enzima Ramificadora de 1,4-alfa-Glucana/química , Amido/química , Amido/metabolismo , Digestão , Sistemas CRISPR-Cas/genética , Amilopectina/química , Amilopectina/metabolismo , Amilose/química , Amilose/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo
5.
Mol Biol Rep ; 51(1): 958, 2024 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-39230778

RESUMO

Sheath blight, caused by the fungus Rhizoctonia solani, is a major problem that significantly impacts rice production and can lead to substantial yield losses. The disease has become increasingly problematic in recent years due to the widespread use of high-yielding semi-dwarf rice cultivars, dense planting, and heavy application of nitrogenous fertilizers. The disease has become more challenging to manage due to its diverse host range and the lack of resistant cultivars. Despite utilizing traditional methods, the problem persists without a satisfactory solution. Therefore, modern approaches, including advanced breeding, transgenic methods, genome editing using CRISPR/Cas9 technology, and nanotechnological interventions, are being explored to develop rice plants resistant to sheath blight disease. This review primarily focuses on these recent advancements in combating the sheath blight disease.


Assuntos
Biotecnologia , Sistemas CRISPR-Cas , Resistência à Doença , Edição de Genes , Oryza , Melhoramento Vegetal , Doenças das Plantas , Rhizoctonia , Oryza/genética , Oryza/microbiologia , Doenças das Plantas/microbiologia , Doenças das Plantas/genética , Resistência à Doença/genética , Rhizoctonia/patogenicidade , Melhoramento Vegetal/métodos , Edição de Genes/métodos , Sistemas CRISPR-Cas/genética , Biotecnologia/métodos , Plantas Geneticamente Modificadas/genética , Nanotecnologia/métodos
6.
Prog Mol Biol Transl Sci ; 208: 161-183, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266181

RESUMO

Cell and gene therapy are innovative biomedical strategies aimed at addressing diseases at their genetic origins. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) systems have become a groundbreaking tool in cell and gene therapy, offering unprecedented precision and versatility in genome editing. This chapter explores the role of CRISPR in gene editing, tracing its historical development and discussing biomolecular formats such as plasmid, RNA, and protein-based approaches. Next, we discuss CRISPR delivery methods, including viral and non-viral vectors, followed by examining the various engineered CRISPR variants for their potential in gene therapy. Finally, we outline emerging clinical applications, highlighting the advancements in CRISPR for breakthrough medical treatments.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Terapia Genética , Sistemas CRISPR-Cas/genética , Humanos , Terapia Genética/métodos , Edição de Genes/métodos , Animais , Terapia Baseada em Transplante de Células e Tecidos/métodos
7.
Prog Mol Biol Transl Sci ; 208: 19-41, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266183

RESUMO

Prokaryotic adaptive immune systems called CRISPR-Cas systems have transformed genome editing by allowing for precise genetic alterations through targeted DNA cleavage. This system comprises CRISPR-associated genes and repeat-spacer arrays, which generate RNA molecules that guide the cleavage of invading genetic material. CRISPR-Cas is classified into Class 1 (multi-subunit effectors) and Class 2 (single multi-domain effectors). Its applications span combating antimicrobial resistance (AMR), targeting antibiotic resistance genes (ARGs), resensitizing bacteria to antibiotics, and preventing horizontal gene transfer (HGT). CRISPR-Cas3, for example, effectively degrades plasmids carrying resistance genes, providing a precise method to disarm bacteria. In the context of ESKAPE pathogens, CRISPR technology can resensitize bacteria to antibiotics by targeting specific resistance genes. Furthermore, in tuberculosis (TB) research, CRISPR-based tools enhance diagnostic accuracy and facilitate precise genetic modifications for studying Mycobacterium tuberculosis. CRISPR-based diagnostics, leveraging Cas endonucleases' collateral cleavage activity, offer highly sensitive pathogen detection. These advancements underscore CRISPR's transformative potential in addressing AMR and enhancing infectious disease management.


Assuntos
Infecções Bacterianas , Sistemas CRISPR-Cas , Sistemas CRISPR-Cas/genética , Humanos , Infecções Bacterianas/microbiologia , Infecções Bacterianas/genética , Edição de Genes , Bactérias/genética
8.
Prog Mol Biol Transl Sci ; 208: 1-17, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266179

RESUMO

Genome editing involves altering of the DNA in organisms including bacteria, plants, and animals using molecular scissors that helps in treatment and diagnosis of various diseases. Genome editing technology is exponentially growing and have been developed for enabling precise genomic alterations and the addition, removal, and correction of genes. These modifications begin with the creation of double-stranded breaks (DSBs) that is generated by nucleases and can be joined through homology-directed repair (HDR) or non-homologous end-joining (NHEJ). NHEJ is quick but increases mutation chances due to deletions and insertions of nucleotides at the break site, while HDR uses homologous templates for precise repair and targeted DNA specific to the gene or sequence. Other methods such as zinc-finger protein is a transcription factor that binds with DNA and binds specific to that sequence, which uniquely recognise 3-base pairs of DNA. TALENs consists of two domains: TALE domain, a transcription activator and FokI that is a restriction endonuclease that cuts the DNA at specific sites. CRISPR-Cas systems are clustered regularly interspersed short palindromic repeats present in various bacterial species. These sequences activate RNA-guided DNA cleavage, aiding in the development of an adaptive immune defence against foreign DNA. CRISPR-Cas9 is widely used for genome editing, regulation, diagnostic and many.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Sistemas CRISPR-Cas/genética , Humanos , Animais
9.
Prog Mol Biol Transl Sci ; 208: 211-229, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266184

RESUMO

Cancer has been a primary contributor to morbidity and mortality worldwide. With an increasing trend of incidence and prevalence of cancer, progress has also been made in its treatment, starting from radiation and chemotherapy to immunotherapy and gene therapy. CRISPR-Cas technique, a promising gene editing tool, has been employed in cancer research for novel treatment regimens, identification of therapeutic targets, and unraveling the genetic mechanisms behind oncogenesis. CRISPR-based genome editing helped in identifying the roles of specific genetic factors linked to treatment resistance, metastasis, and cancer development. CRISPR allows the discovery of genes and treatment options through specifically interrupting tumor activators or activating tumor suppressor genes in cancer cells. Advancements in CRISPR technology, especially the use of immune cells like chimeric antigen receptor (CAR) T cells, has the potential to revolutionize personalized cancer treatment by precisely targeting and killing cancer cells. Furthermore, reactivating tumor suppressor genes makes cancer cells more susceptible to chemotherapy or immunotherapy. CRISPR-mediated genome editing can, hence, help to overcome resistance to traditional cancer treatments. The current manuscript covers that how is the CRISPR technology propelling revolutionary development in the field of cancer research, providing advance perspectives on the molecular causes of the disease and creating new lines for the development of more precise and potent cancer therapies.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Neoplasias , Humanos , Sistemas CRISPR-Cas/genética , Neoplasias/genética , Neoplasias/terapia , Animais
10.
Prog Mol Biol Transl Sci ; 208: 231-259, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266185

RESUMO

A body develops an autoimmune illness when its immune system mistakenly targets healthy cells and organs. Eight million people are affected by more than 80 autoimmune diseases. The public's and individuals' well-being is put at risk. Type 1 diabetes, lupus, rheumatoid arthritis, and multiple sclerosisare autoimmune diseases. Tissue injury, nociceptive responses, and persistent inflammation are the results of these stresses. Concerns about healthcare costs, health, and physical limitations contribute to these issues. Given their prevalence, it is crucial to enhance our knowledge, conduct thorough research, and provide all-encompassing support to women dealing with autoimmune diseases. This will lead to better public health and better patient outcomes. Most bacteria's immune systems employ CRISPR-Cas, a state-of-the-art technique for editing genes. For Cas to break DNA with pinpoint accuracy, a guide RNA employs a predetermined enzymatic pathway. Genetic modifications started. After it was developed, this method was subjected to much research on autoimmune diseases. By modifying immune pathways, CRISPR gene editing can alleviate symptoms, promote immune system tolerance, and decrease autoimmune reactivity. The autoimmune diseases that CRISPR-Cas9 targets now have no treatment or cure. Results from early clinical trials and preclinical studies of autoimmune medicines engineered using CRISPR showed promise. Modern treatments for rheumatoid arthritis,multiple sclerosis, and type 1 diabetes aim to alter specific genetic or immune mechanisms. Accurate CRISPR editing can fix autoimmune genetic disorders. Modifying effector cells with CRISPR can decrease autoimmune reactions. These cells include cytotoxic T and B lymphocytes. Because of improvements in delivery techniques and kits, CRISPR medications are now safer, more effective, and more accurately targeted. It all comes down to intricate immunological reactions and unexpected side consequences. Revolutionary cures for autoimmune problems and highly personalized medical therapies have been made possible by recent advancements in CRISPR.


Assuntos
Doenças Autoimunes , Sistemas CRISPR-Cas , Humanos , Sistemas CRISPR-Cas/genética , Doenças Autoimunes/genética , Doenças Autoimunes/terapia , Doenças Autoimunes/imunologia , Animais , Edição de Genes , Terapia Genética/métodos
11.
Prog Mol Biol Transl Sci ; 208: 109-160, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266180

RESUMO

Protozoan parasitic diseases pose a substantial global health burden. Understanding the pathogenesis of these diseases is crucial for developing intervention strategies in the form of vaccine and drugs. Manipulating the parasite's genome is essential for gaining insights into its fundamental biology. Traditional genomic manipulation methods rely on stochastic homologous recombination events, which necessitates months of maintaining the cultured parasites under drug pressure to generate desired transgenics. The introduction of mega-nucleases (MNs), zinc-finger nucleases (ZFNs), and transcription activator-like effector nucleases (TALENs) greatly reduced the time required for obtaining a desired modification. However, there is a complexity associated with the design of these nucleases. CRISPR (Clustered regularly interspaced short palindromic repeats)/Cas (CRISPR associated proteins) is the latest gene editing tool that provides an efficient and convenient method for precise genomic manipulations in protozoan parasites. In this chapter, we have elaborated various strategies that have been adopted for the use of CRISPR-Cas9 system in Plasmodium, Leishmania and Trypanosoma. We have also discussed various applications of CRISPR-Cas9 pertaining to understanding of the parasite biology, development of drug resistance mechanism, gene drive and diagnosis of the infection.


Assuntos
Sistemas CRISPR-Cas , Infecções por Protozoários , Sistemas CRISPR-Cas/genética , Humanos , Infecções por Protozoários/genética , Infecções por Protozoários/parasitologia , Animais , Edição de Genes
12.
Prog Mol Biol Transl Sci ; 208: 261-284, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266186

RESUMO

CRISPR-Cas systems have revolutionised precision medicine by enabling personalised treatments tailored to an individual's genetic profile. Various CRISPR technologies have been developed to target specific disease-causing genes in blood cancers, and some have advanced to clinical trials. Although some studies have explored the in vivo applications of CRISPR-Cas systems, several challenges continue to impede their widespread use. Furthermore, CRISPR-Cas technology has shown promise in improving the response of immunotherapies to blood cancers. The emergence of CAR-T cell therapy has shown considerable success in the targeting and correcting of disease-causing genes in blood cancers. Despite the promising potential of CRISPR-Cas in the treatment of blood cancers, issues related to safety, ethics, and regulatory approval remain significant hurdles. This comprehensive review highlights the transformative potential of CRISPR-Cas technology to revolutionise blood cancer therapy.


Assuntos
Sistemas CRISPR-Cas , Neoplasias Hematológicas , Humanos , Sistemas CRISPR-Cas/genética , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/genética , Animais , Edição de Genes/métodos
13.
Prog Mol Biol Transl Sci ; 208: 43-58, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266187

RESUMO

Clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system possess a broad range of applications for genetic modification, diagnosis and treatment of infectious as well as non-infectious disease. The CRISPR-Cas system is found in bacteria and archaea that possess the Cas protein and guide RNA (gRNA). Cas9 and gRNA forms a complex to target and cleave the desired gene, providing defense against viral infections. Human immunodeficiency virus (HIV), hepatitis B virus (HBV), herpesviruses, human papillomavirus (HPV), and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) cause major life threatening diseases which cannot cure completely by drugs. This chapter describes the present strategy of CRISPR-Cas systems for altering the genomes of viruses, mostly human ones, in order to control infections.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Humanos , Sistemas CRISPR-Cas/genética , Viroses/genética , Viroses/terapia , Viroses/virologia , Vírus/genética , Genoma Viral/genética
14.
Prog Mol Biol Transl Sci ; 208: 185-209, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266182

RESUMO

The CRISPR-Cas9 method has revolutionized the gene editing. Epigenetic changes, including DNA methylation, RNA modification, and changes in histone proteins, have been intensively studied and found to play a key role in the pathogenesis of human diseases. CRISPR-While the utility of DNA and chromatin modifications, known as epigenetics, is well understood, the functional significance of various alterations of RNA nucleotides has recently gained attention. Recent advancements in improving CRISPR-based epigenetic modifications has resulted in the availability of a powerful source that can selectively modify DNA, allowing for the maintenance of epigenetic memory over several cell divisions. Accurate identification of DNA methylation at specific locations is crucial for the prompt detection of cancer and other diseases, as DNA methylation is strongly correlated to the onset as well as the advancement of such conditions. Genetic or epigenetic perturbations can disrupt the regulation of imprinted genes, resulting in the development of diseases. When histone code editors and DNA de-/ methyltransferases are coupled with catalytically inactive Cas9 (dCas9), and CRISPRa and CRISPRi, they demonstrate excellent efficacy in editing the epigenome of eukaryotic cells. Advancing and optimizing the extracellular delivery platform can, hence, further facilitate the manipulation of CRISPR-Cas9 gene editing technique in upcoming clinical studies. The current chapter focuses on how the CRISP/ Cas9 system provides an avenue for the epigenetic modifications and its employability for human benefit.


Assuntos
Sistemas CRISPR-Cas , Epigênese Genética , Humanos , Sistemas CRISPR-Cas/genética , Animais , Edição de Genes/métodos , Metilação de DNA/genética
15.
Prog Mol Biol Transl Sci ; 208: 59-81, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266188

RESUMO

CRISPR-Cas technology has revolutionized microbiome research by enabling precise genetic manipulation of microbial communities. This review explores its diverse applications in gut microbiome studies, probiotic development, microbiome diagnostics, pathogen targeting, and microbial community engineering. Engineered bacteriophages and conjugative probiotics exemplify CRISPR-Cas's capability for targeted bacterial manipulation, offering promising strategies against antibiotic-resistant infections and other gut-related disorders. CRISPR-Cas systems also enhance probiotic efficacy by improving stress tolerance and colonization in the gastrointestinal tract. CRISPR-based techniques in diagnostics enable early intervention by enabling fast and sensitive pathogen identification. Furthermore, CRISPR-mediated gene editing allows tailored modification of microbial populations, mitigating risks associated with horizontal gene transfer and enhancing environmental and health outcomes. Despite its transformative potential, ethical and regulatory challenges loom large, demanding robust frameworks to guide its responsible application. This chapter highlights CRISPR-Cas's pivotal role in advancing microbiome research toward personalized medicine and microbial therapeutics while emphasizing the imperative of balanced ethical deliberations and comprehensive regulatory oversight.


Assuntos
Sistemas CRISPR-Cas , Microbioma Gastrointestinal , Sistemas CRISPR-Cas/genética , Humanos , Microbioma Gastrointestinal/genética , Animais , Edição de Genes , Probióticos/uso terapêutico
16.
Prog Mol Biol Transl Sci ; 208: 83-107, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39266189

RESUMO

Fungi contain a wide range of bioactive secondary metabolites (SMs) that have numerous applications in various fields, including agriculture, medicine, human health, and more. It is common for genes responsible for the production of secondary metabolites (SMs) to form biosynthetic gene clusters (BGCs). The identification and analysis of numerous unexplored gene clusters (BGCs) and their corresponding substances (SMs) has been significantly facilitated by the recent advancements in genomic and genetic technologies. Nevertheless, the exploration of secondary metabolites with commercial value is impeded by a variety of challenges. The emergence of modern CRISPR/Cas technologies has brought about a paradigm shift in fungal genetic engineering, significantly streamlining the process of discovering new bioactive compounds. This study begins with an examination of fungal biosynthetic gene clusters (BGCs) and their interconnections with the secondary metabolites (SMs) they generate. Following that, a brief summary of the conventional methods employed in fungal genetic engineering is provided. This study explores various sophisticated CRISPR/Cas-based methodologies and their utilization in examining the synthesis of secondary metabolites (SMs) in fungi. The chapter provides an in-depth analysis of the limitations and obstacles encountered in CRISPR/Cas-based systems when applied to fungal genetic engineering. It also proposes promising avenues for future research to optimize the efficiency of these systems.


Assuntos
Sistemas CRISPR-Cas , Fungos , Micoses , Sistemas CRISPR-Cas/genética , Micoses/genética , Micoses/microbiologia , Humanos , Fungos/genética , Família Multigênica , Engenharia Genética
17.
Theranostics ; 14(12): 4894-4915, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39239522

RESUMO

Rationale: Regulatory processes of transcription factors (TFs) shape heart development and influence the adult heart's response to stress, contributing to cardiac disorders. Despite their significance, the precise mechanisms underpinning TF-mediated regulation remain elusive. Here, we identify that EBF1, as a TF, is highly expressed in human heart tissues. EBF1 is reported to be associated with human cardiovascular disease, but its roles are unclear in heart. In this study, we investigated EBF1 function in cardiac system. Methods: RNA-seq was utilized to profile EBF1 expression patterns. CRISPR/Cas9 was utilized to knock out EBF1 to investigate its effects. Human pluripotent stem cells (hPSCs) differentiated into cardiac lineages were used to mimic cardiac development. Cardiac function was evaluated on mouse model with Ebf1 knockout by using techniques such as echocardiography. RNA-seq was conducted to analyze transcriptional perturbations. ChIP-seq was employed to elucidate EBF1-bound genes and the underlying regulatory mechanisms. Results: EBF1 was expressed in some human and mouse cardiomyocyte. Knockout of EBF1 inhibited cardiac development. ChIP-seq indicated EBF1's binding on promoters of cardiogenic TFs pivotal to cardiac development, facilitating their transcriptional expression and promoting cardiac development. In mouse, Ebf1 depletion triggered transcriptional perturbations of genes, resulting in cardiac remodeling. Mechanistically, we found that EBF1 directly bound to upstream chromatin regions of cardiac hypertrophy-inducing genes, contributing to cardiac hypertrophy. Conclusions: We uncover the mechanisms underlying EBF1-mediated regulatory processes, shedding light on cardiac development, and the pathogenesis of cardiac remodeling. These findings emphasize EBF1's critical role in orchestrating diverse aspects of cardiac processes and provide a promising therapeutic intervention for cardiomyopathy.


Assuntos
Perfilação da Expressão Gênica , Miócitos Cardíacos , Transativadores , Animais , Humanos , Camundongos , Transativadores/genética , Transativadores/metabolismo , Miócitos Cardíacos/metabolismo , Diferenciação Celular/genética , Coração/fisiopatologia , Camundongos Knockout , Células-Tronco Pluripotentes/metabolismo , Transcriptoma/genética , Sistemas CRISPR-Cas/genética
18.
Cell ; 187(19): 5220-5222, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39303688

RESUMO

Fanzors are recently characterized RNA-guided DNA endonucleases found in eukaryotic organisms. In this issue of Cell, Xu, Saito et al. reveal the structural diversity of Fanzors and identify key features shared with TnpB and Cas12 proteins, providing a comprehensive perspective on their molecular function and evolution.


Assuntos
Sistemas CRISPR-Cas , Sistemas CRISPR-Cas/genética , Eucariotos/genética , RNA Guia de Sistemas CRISPR-Cas/genética , RNA Guia de Sistemas CRISPR-Cas/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Proteínas Associadas a CRISPR/genética , DNA/genética , DNA/metabolismo , Endodesoxirribonucleases/metabolismo , Endodesoxirribonucleases/genética , Humanos
19.
Mol Plant Pathol ; 25(10): e70010, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39344009

RESUMO

Cassava starch is a widely used raw material for industrial production and food source for people. However, cassava bacterial blight (CBB) caused by Xanthomonas axonopodis pv. manihotis (Xam) results in severe yield losses and is the most destructive bacterial disease in all worldwide cassava-growing regions. Xam11 is a highly pathogenic subspecies from China that infects the Chinese local cassava South China No. 8 (SC8) cultivar with marked symptoms. This study showed that the transcription activator-like effector TALE20Xam11 of Xam11 strain regulates the expression of disease-susceptibility gene MeSWEET10a by binding to the EBETALE20 region of the MeSWEET10a promoter in cassava cultivar SC8. CRISPR/Cas9-generated mutations of the EBETALE20 region resulted in a significant reduction in MeSWEET10a expression after infection by Xam11, correlating with reduced disease symptoms, smaller lesion sizes and decreased bacterial proliferation compared with the wild type. Importantly, the edited plants maintained normal growth, development and yield characteristics under greenhouse conditions. The results lay a research foundation for breeding resistant cassava cultivar SC8 to bacterial blight.


Assuntos
Resistência à Doença , Manihot , Doenças das Plantas , Regiões Promotoras Genéticas , Manihot/microbiologia , Manihot/genética , Doenças das Plantas/microbiologia , Doenças das Plantas/genética , Doenças das Plantas/imunologia , Resistência à Doença/genética , Regiões Promotoras Genéticas/genética , Xanthomonas axonopodis/patogenicidade , Edição de Genes , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Sistemas CRISPR-Cas/genética , Plantas Geneticamente Modificadas , Regulação da Expressão Gênica de Plantas
20.
Biotechnol J ; 19(9): e2400309, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39295562

RESUMO

The filamentous fungus Rhizopus oryzae is one of the main industrial strains for the production of a series of important chemicals such as ethanol, lactic acid, and fumaric acid. However, the lack of efficient gene editing tools suitable for R. oryzae makes it difficult to apply technical methods such as metabolic engineering regulation and synthetic biology modification. A CRISPR-Cas9 system suitable for efficient genome editing in R. oryzae was developed. Firstly, four endogenous U6 promoters of R. oryzae were identified and screened with the highest transcriptional activity for application to sgRNA transcription. It was then determined that the U6 promoter mediated CRISPR/Cas9 system has the ability to efficiently edit the genome of R. oryzae through NHEJ and HDR-mediated events. Furthermore, the newly constructed CRISPR-Cas9 dual sgRNAs system can simultaneously disrupt or insert different fragments of the R. oryzae genome. Finally, this CRISPR-Cas9 system was applied to the genome editing of R. oryzae by knocking out pyruvate carboxylase gene (PYC) and pyruvate decarboxylase gene (pdcA) and knocking in phosphofructokinase (pfkB) from Escherichia coli and L-lactate dehydrogenase (L-LDH) from Heyndrickxia coagulans, which resulted in a substantial increase in L-LA production. In summary, this study showed that the CRISPR/Cas9-based genome editing tool is efficient for manipulating genes in R. oryzae.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Ácido Láctico , Engenharia Metabólica , Rhizopus oryzae , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Ácido Láctico/metabolismo , Engenharia Metabólica/métodos , Rhizopus oryzae/genética , L-Lactato Desidrogenase/genética , L-Lactato Desidrogenase/metabolismo , Genoma Fúngico/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Piruvato Descarboxilase/genética , Piruvato Descarboxilase/metabolismo , Rhizopus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA