• Account
  • Account
    Inquiries
    0

    Inquired items

    0items total Subtotal:$ 0
  • Cart
    Cart

    Inquired items

    items total Subtotal:$ 0

All Departments

  • Single-Cell RNA Sequencing
    Introduction Single-cell RNA sequencing (scRNA-seq) provides the expression profile of individual cells. Through genes clustering analyses, rare cell types within a cell population can be identified, thereby making characterization of the subpopulation structure of a heterogeneous cell population become available. Single-cell RNA sequencing on numerous single cells can identify uncommon RNAs such as the RNA with low copy number, which may exert important functions in the cells, and also reveal the copy-number distribution of the whole mRNA population in individual cells.  Despite the advances in sequencing technologies, it is still unattainable to sequence RNA directly from single cells. Thus, in the current scRNA-seq protocols, RNA still needs to be converted to cDNA for sequencing. Principally, the current scRNA-seq methods contain the following steps: isolation of single-cell and RNA, reverse transcription (RT), amplification, library generation and sequencing.
  • Single Cell Genome (DNA)Sequencing
    Introduction Single cell DNA genome sequencing involves isolating a single cell, performing whole-genome-amplification (WGA), constructing sequencing libraries and then sequencing the DNA using a next-generation sequencer. One popular method used for single cell genome sequencing is multiple displacement amplification (MDA), and this enables research into various areas such as microbial genetics, ecology and infectious diseases.  Single cell genomics is the one way to identify microbiomes’ identities and its genomes, and cancer sequencing is also an emerging application of scDNAseq. Fresh or frozen tumors may be analyzed and categorized with respect to SCNAs, SNVs, and rearrangements quite well using whole genome DNAS approaches. Cancer scDNAseq is particularly useful for examining the depth of complexity and compound mutations present in amplified therapeutic targets such as receptor tyrosine kinase genes (EGFR, PDGFRA, etc.) where conventional population-level approaches of the bulk tumor are not able to resolve the co-occurrence patterns of these mutations within single cells of the tumor. 
  • Targeted Gene Sequencing
    Introduction Targeted gene sequencing panels are useful tools for analyzing specific mutations in a given sample. With targeted resequencing, a subset of genes or regions of the genome are isolated and sequenced. Targeted approaches using NGS allow researchers to focus time, expenses, and data analysis on specific areas of interest. Such targeted analysis can include the exome, specific genes of interest (custom content), targets within genes, or mitochondrial DNA. Targeted gene sequencing also produces a smaller, more manageable data set compared to broader approaches such as whole-genome sequencing, making analysis easier. There are two methods for the targeted gene sequencing: target enrichment and amplicon generation. In the target enrichment method, Regions of interest are captured by hybridization to biotinylated probes and then isolated by magnetic pulldown, while in amplicon generation, regions of interest are amplified and purified using highly multiplexed oligo pools.  The difference between targeted sequencing and whole-genome sequencing is like below:
  • Bacterial Genome Sequencing
    Introduction Next-generation sequencing technologies made rapid assembly of draft microbial genomes possible, and sequencing the entire microbial genome is important for generating accurate reference genomes, for microbial identification, and other comparative genomic studies. Unlike traditional methods, NGS-based microbial genome sequencing doesn’t rely on labor-intensive cloning steps, saving time and simplifying the workflow. NGS can identify low-frequency variants and genome rearrangements that may be missed or are too expensive to identify using other methods. The high-throughput sequencing is one of the NGS methods that we could use for the bacterial genome sequencing, including:  (1). identification of target DNA sequences and antigens to rapidly develop diagnostic tools;  (2). precise strain identification for epidemiological typing and pathogen monitoring during outbreaks; (3). investigation of strain properties, such as the presence of antibiotic resistance or virulence factors. The procedures are as follows:
  • smallRNA/microRNA/circRNA/LncRNA Sequencing
    smallRNA/microRNA Sequencing Small non-coding RNAs act in gene silencing and post-transcriptional regulation of gene expression. Small RNA sequencing is a technique to isolate and sequence small RNA species, such as microRNAs (miRNAs).  With small RNA-Seq, you can discover novel miRNAs and other small non-coding RNAs, and examine the differential expression of all small RNAs in any sample. You can characterize variations such as isomiRs with single-base resolution, as well as analyze any small RNA or miRNA without prior sequence or secondary structure information. The high throughput sequencing technique provides high sensitivity and specificity to analyze the abundance of sRNA/miRNA sequence in a sample as well as to discover novel microRNA species. circRNA sequencing Circular RNAs (CircRNAs) are a recent novel class of abundant, stable and ubiquitous RNAs addition to the growing list of types of non-coding RNA molecules, they can arise from both exons (exonic circRNA) and introns (intronic circRNA) and serve as miRNA or RNA-binding protein ‘sponges’, sequestering miRNAs and preventing their interactions wit...
  • Whole Transcriptome Shotgun Sequencing
    Introduction Whole Transcriptome Shotgun Sequencing (WTSS), also called RNA sequencing, uses next-generation sequencing (NGS) to reveal the presence and quantity of RNA in a biological sample at a given moment in time.  WTSS including a wide variety of applications from simple mRNA profiling to discovery and analysis of the entire transcriptome, including both coding mRNA and non-coding RNA (e.g., miRNA, small RNAs, linc RNAs). These applications are extremely popular for NGS platforms as they uncover information that may be missed by array-based platforms, as no prior knowledge of the transcript sequence is needed. Also, as it is sequencing-based, it is well suited for specialty applications such as RNA editing and allele-specific expression. While there are a variety of RNA-Seq applications and protocols, most follow the basic strategy of isolating RNA (such as with poly dT to pull down mRNA), converting it to DNA and then adding adaptor sequences to generate a library suitable for sequencing. The WTSS procedures are as follows:
  • Whole Exome Sequencing
    Introduction The whole-exome sequencing (WES) is a genomic technique for sequencing all of the protein-coding genes in a genome (known as the exome). It consists of two steps: the first step is to select only the subset of DNA that encodes proteins. And the second step is to sequence the exonic DNA using any high-throughput DNA sequencing technology. Exome sequencing can efficiently identify coding variants across a wide range of applications, including population genetics, genetic disease, and cancer studies. The WES advantages are:  (1). Identifies variants across a wide range of applications.  (2). Achieves comprehensive coverage of coding regions.  (3). Provides a cost-effective alternative to whole-genome sequencing.  (4). Produces a smaller, more manageable data set for faster, easier analysis compared to whole-genome approaches.
  • Plant and Animal Whole Genome Re-Sequencing
    Introduction Plant and animal whole-genome re-sequencing involves sequencing the entire genome of a plant or animal, and comparing the sequence to that of a known reference genome. Re-sequencing of the plant and animal genome will identify genetic variations such as SNPs and InDels, and discover other genetic changes of the sequenced species.  It has been applied for the identification of functional genes and markers of important traits to facilitate molecular breeding and to improve agricultural production and conservation. The resequencing process is as follows:
  • Plant and Animal Gene Sequencing
    Introduction De Novo Sequencing is one of the methods for plant and animal genome sequencing, the name De Novo refers to sequencing a novel genome where there is no reference sequence available for alignment. Sequence reads are assembled as contigs, and the coverage quality of de novo sequence data depends on the size and continuity of the contigs. De Novo Sequencing has many advantages compared to other sequencing methods: First, it generates accurate reference sequences, even for complex or polyploid genomes. Second, it provides useful information for mapping genomes of novel organisms or finishing genomes of known organisms. Third, it clarifies highly similar or repetitive regions for accurate de novo assembly, and last, it identifies structural variants and complex rearrangements, such as deletions, inversions, or translocations.
  • NGS Based Service
    Introduction Next-generation sequencing (NGS), also known as high-throughput sequencing, is the catch-all term used to describe a number of different modern sequencing technologies including Illumina (Solexa) sequencing, Roche 454 sequencing, Ion torrent: Proton / PGM sequencing, and SOLiD sequencing. These recent technologies allow us to sequence DNA and RNA much more quickly and cheaply than the previously used Sanger sequencing, and as such have revolutionized the study of genomics and molecular biology.
Home Previous 1 2 3 Next Last 26 records total, 3 pages To page
Contact Us