Life science Research

Life science Research

about images

Antibodies & Assay-Kits

Antibodies are glycoproteins found in body fluids including blood, milk, and mucous secretions and serve an essential role in the immune system that protects animals from infection or the cytotoxic effects of foreign compounds. Antibodies will bind with high affinity to an invasive molecule. Normally the binding is to only part of a large molecule (the epitope) and so there may be many different antibodies for a particular compound.

Antibodies have become essential tools for biological research because of their very specific recognition and affinity for one compound (the antigen). This has not only led to the use of antibodies in the recognition of specific cellular components but also to the development of routine diagnostic medical tests. More recently antibodies have been used as therapeutic agents for the treatment of human disease.

View More

Cell Culture & Analysis

Cell culture is fundamental in life science research and development Whether your goal is to establish relevant cell models to probe complex biology, establish drug discovery and drug development assays, or produce recombinant proteins or therapeutics.

Our cell culture workflow portfolio includes advanced Cell culture solutions, Cell counting and imaging equipment, culture ware, extracellular matrices, hydrogels, sterile filtration consumables, reagents, media, sera, and growth factors for 2D experiments.

Stem cells are undifferentiated, or “blank,” cells. This means they’re capable of developing into cells that serve numerous functions in different parts of the body. Most cells in the body are differentiated cells. These cells can only serve a specific purpose in a particular organ. For example, red blood cells are specifically designed to carry oxygen through the blood

Stem cells are cells that haven’t differentiated yet. They have the ability to divide and make an indefinite number of copies of themselves. Other cells in the body can only replicate a limited number of times before they begin to break down. When a stem cell divides, it can either remain a stem cell or turn into a differentiated cell, such as a muscle cell or a red blood cell. Since stem cells have the ability to turn into various other types of cells, scientists believe that they can be useful for treating and understanding diseases. Thus, stem cells have widened horizon of scope of research potential and therapies too.

Recently , Three-dimensional (3D) cell culture systems have gained increasing interest in drug discovery and tissue engineering due to their evident advantages in providing more physiologically relevant information and more predictive data for in vivo tests. The innovations and development in 3D culture systems for drug discovery over the past 5 years are also reviewed in the article, emphasizing the cellular response to different classes of anticancer drugs, focusing particularly on similarities and differences between 3D and 2D models across the field. The progression and advancement in the application of 3D cell cultures in cell-based biosensors .

3D cell culture is an in-vitro technique wherein the cells can grow in an artificially created environment. These environments closely resemble the architecture and functioning of the native tissue. 3D cell culture technique helps stimulate cell differentiation, proliferation, and migration by interacting with their three-dimensional surroundings as they would in the in-vivo environment. As 3D cell cultures can mimic the structure, activity, and microenvironment of the in-vivo tissues, this technique has varied applications in the fields of drug screening, regenerative medicine, stem cell therapies, cancer research and cell biology. The extracellular matrix in 3D cell cultures enables cell–cell communication by direct contact as in in-vivo environment by secreting cytokines and trophic factors. These factors are changed in a 2D environment that can significantly affect the cell–cell communication, which in turn can alter the cell morphology and proliferation. As 2D cultures cannot recapitulate the architecture and complex cellular matrices as in 3D cultures, this technique is gaining popularity in healthcare research sector. In addition, 3D cell cultures can provide results with improved efficiency and reduce the cost of overall R&D process.

Broadly, 3D cell culture techniques are classified as Scaffold-based or non-scaffold-based techniques.

about images

In scaffold based techniques cells are grown in presence of a support. 2 major types of support can be used:

1. Hydrogel-based support: Hydrogels are by definition polymer networks extensively swollen with water. Cells can be embedded in these hydrogels or simply coated at the surface. Depending on the nature of the polymer, hydrogels can be classified in to different categories (ECM protein-based hydrogels, natural hydrogels and synthetic hydrogels) with distinct properties.

2. Polymeric hard material based support: cells are cultivated in presence of fibers or sponge-like structures: cell recover a more physiological shape because they are not plated on a flat surface. Materials used for these supports can be polystyrene (adapted for imaging studies because of its transparency) but also biodegradable tools like polycaprolactone.

Scaffold-free techniques allow the cells to self-assemble to form non-adherent cell aggregates called spheroids. Spheroids mimic the solid tissues by secreting their own extracellular matrix and displaying differential nutrient availability. Spheroids grown via non-scaffold based techniques are consistent in size and shape and are better in-vitro cellular models for high-throughput screening The evolution of 3D cell culture has the potential to bridge the gap between in-vitro and in vivo experiments. The convenience of handling cells in-vitro while obtaining results that reflect in-vivo condition and avoiding ethical concerns of animal usage is making 3D cell culture techniques increasingly popular among researchers, but choosing the right system to develop a 3D cell culture model is not a trivial question.

about images
View More

Flow Cytometry

Flow cytometry is a powerful tool because it allows simultaneous multiparametric analysis of the physical and chemical characteristics of up to thousands of particles per second. This makes it a rapid and quantitative method for analysis and purification of cells in suspension. Using flow, we can determine the phenotype and function and even sort live cells.

FACS is an abbreviation for fluorescence-activated cell sorting, which is a flow cytometry technique that further adds a degree of functionality. By utilizing highly specific antibodies labelled with fluorescent conjugates, FACS analysis allows us to simultaneously collect data on, and sort a biological sample by a nearly limitless number of different parameters.


about images
View More

Synthetic Biology

Synthetic biology is a new interdisciplinary area that involves the application of engineering principles to biology. It aims at the (re-)design and fabrication of biological components and systems that do not already exist in the natural world. Synthetic biology combines chemical synthesis of DNA with growing knowledge of genomics to enable researchers to quickly manufacture catalogued DNA sequences and assemble them into new genomes.

Improvements in the speed and cost of DNA synthesis are enabling scientists to design and synthesize modified bacterial chromosomes that can be used in the production of advanced biofuels, bio-products, renewable chemicals, bio-based specialty chemicals (pharmaceutical intermediates, fine chemicals, food ingredients), and in the health care sector as well.


about images
View More

Cell & Gene Therapy

In recent years, Cell and Gene Therapies have started to become a reality with innovative new treatments for diseases being developed. These therapies may have the potential to transform patients’ lives and cure diseases in one or only a few treatments by addressing the root cause of the diseases rather than the symptoms.

Cell and Gene Therapy involves extracting cells, protein or DNA from a patient or donor and altering the genetics to provide a targeted result once reinjected into the patient. Within the past 50 years significant advances in biopharmaceuticals have resulted in significant successes in the treatment of a wide range of haematological, neurological and oncological disorders. Overall, this method of therapy can offer a more effective and longer lasting effect than traditional medicines.

This technique of altering genetic material can greatly impact an individual’s quality of life and offer a sense of hope when more traditional methodologies may have failed them or are not available. There are a variety of innovative methods that can be utilised in order to obtain the desired results.

Cell Therapy can be either from a patient’s own body (autologous) or from a donor (allogenic) and have the potential for unlimited application when treating disease. A variety of cells can be utilised in the procedure of Cell Therapy ranging from lymphocytes, red and white blood cells, platelets and most notoriously stem cells.
Pluripotent stem cells are capable of the production of any human cell, operating as a potential source for inaccessible or scarce cells currently present in the patient. These cells can be divided into two forms, embryonic stem cells (ESCs) and induced pluripotent stem cells (iPCSs).
ESCs – Derived from early-stage embryos (blastocysts) and can develop into over 200 human cell types if coded to do so. Another key factor is their capability to replicate indefinitely.

iPCSs – Mature somatic cells, commonly skin or blood cells that have been reprogrammed into an ESC like condition, it additionally possesses the same, critical, infinite replication characteristic as EPCs.

Altering the genetic material can result in the prevention of disease via modifying how a single protein, or group of proteins, is synthesised by the cell. A primary example of this treatment is either substituting a mutated gene with a functional copy or introducing a required gene to the patient. These methods can result in reducing levels of a disease-causing version of a protein, increase effectiveness of the patient’s immune system and/or produce new or modified proteins.

Gene Therapy can be carried out both in vivo (the gene can be delivered directly to the patient’s targeted cells) or ex vivo (where the therapeutic gene is inserted into cells external from the body in a laboratory, before being introduced to the patient). Therapeutic, altered genes are administered via a deactivated virus, most commonly lentiviruses, retroviruses or adeno-associated viruses.

There are a plethora of methods and approaches that can be utilised to execute ex vivo Gene Therapy such as T-Cell Therapy, natural killer Cell Therapy and tumour infiltrating lymphocytes. However, this technique is most frequently applied to hematopoietic stem cells (HSCs) and is utilised to treat immunological and genetic diseases that influence tissues and organs reached by blood cells.

about images

Scaffold-free techniques allow the cells to self-assemble to form non-adherent cell aggregates called spheroids. Spheroids mimic the solid tissues by secreting their own extracellular matrix and displaying differential nutrient availability. Spheroids grown via non-scaffold based techniques are consistent in size and shape and are better in-vitro cellular models for high-throughput screening The evolution of 3D cell culture has the potential to bridge the gap between in-vitro and in vivo experiments. The convenience of handling cells in-vitro while obtaining results that reflect in-vivo condition and avoiding ethical concerns of animal usage is making 3D cell culture techniques increasingly popular among researchers, but choosing the right system to develop a 3D cell culture model is not a trivial question.

about images
View More

DNA & RNA Extractions

Extraction of DNA and RNA is a basic method used in molecular biology. The need for high-quality, highly pure nucleic acid is important for a wide range of research and clinical applications. Nucleic acid purification is an initial step in many molecular biology and genomic workflows.

DNA and RNA samples are often obtained from crude preparations. Genomic DNA, plasmid DNA, and total RNA can be extracted and purified from a variety of sources including bacterial and mammalian cells, plant tissue, fungal tissue, mammalian tissue, blood, plasma, serum, viruses, buccal and nasal swabs, gel matrices, PCRs, and other enzymatic reactions. Isolation of nucleic acid from these samples often involves the lysis of cell membranes or sample homogenization, followed by the removal of proteins, enzymes, detergents, salts, and lipids.

about images

about images
View More

PCR & Real Time PCR

PCR (polymerase chain reaction) tests are a fast, highly accurate way to diagnose certain infectious diseases and genetic changes. The tests work by finding the DNA or RNA of a pathogen (disease-causing organism) or abnormal cells in a sample.

• DNA is the genetic material that contains instructions and information for all living things.

• RNA is another type of genetic material. It contains information that has been copied from DNA and is involved in making proteins. Most viruses and other pathogens contain DNA or RNA.

Unlike many other tests, PCR tests can find evidence of disease in the earliest stages of infection. Other tests may miss early signs of disease because there aren't enough viruses, bacteria, or other pathogens in the sample, or your body hasn't had enough time to develop an antibody response. Antibodies are proteins made by your immune system to attack foreign substances, such as viruses and bacteria. PCR tests can detect disease when there is only a very small amount of pathogens in your body.

For some applications, qualitative nucleic acid detection is sufficient. Other applications, however, demand a quantitative analysis. Real-time PCR can be used for both qualitative and quantitative analysis; choosing the best method for your application requires a broad knowledge of this technology.

about images

about images

Sanger sequencing, also known as chain-termination sequencing or dideoxy sequencing has been the powerhouse of DNA sequencing since its invention in the 1970s. The process is based on the detection of labelled chain-terminating nucleotides that are incorporated by a DNA polymerase during the replication of a template. The method has been extensively used to advance the field of functional and comparative genomics, evolutionary genetics and complex disease research. Notably, the dideoxy method was employed in sequencing the first human genome in 2002. Because of its suitability for routine validation of cloning experiments and PCR fragments, Sanger sequencing remains a popular technique in many laboratories across the world. Applications - What are the advantages of Sanger sequencing?

Sanger DNA sequencing is widely used for research purposes like:

Targeting smaller genomic regions in a larger number of samples
Sequencing of variable regions
Validating results from next-generation sequencing (NGS) studies
Verifying plasmid sequences, inserts, mutations
HLA typing
Genotyping of microsatellite markers
Identifying single disease-causing genetic variants

about images

The DNA sequencing field did not stop evolving with the successful adaptation of Sanger sequencing. The establishment of next-generation sequencing (NGS) and third-generation sequencing technologies offered substantial benefits compared to the traditional dideoxy method. However, the chain-termination method remains widely used in the sequencing field, because it offers several distinct advantages.

Specifically, Sanger sequencing is preferable over NGS for:

Sequencing of single genes
Cost-efficient sequencing of single samples
Verification sequencing for site-directed mutagenesis or the presence of cloned inserts
In some cases, analysis of longer fragments (~1,000 bp in length)
In some cases, less error-prone than NGS Nevertheless, next-generation sequencing is often considered to be superior to Sanger sequencing, especially for project objectives that require:
Cost-efficient, simultaneous interrogation of more than 100 genes at a time
Finding novel variants by increasing the number of targets sequenced per run
Analysis of samples with low-input DNA
Sequencing of whole genomes, especially microbial genomes

about images
View More

Drug Discovery

The complexity in drug development has increased manifolds over the past 40 years, requiring preclinical phase of drug development, investigational new drug (IND) application, and complete clinical testing before marketing approval from the FDA. The goal is to bring more efficient and safer treatments to the patients as quickly as possible after a thorough medical evaluation.

about images
View More

Drug Delivery Systems

Drug delivery systems describe technologies that carry drugs into or throughout the body. These technologies include the method of delivery, such as a pill that you swallow or a vaccine that is injected. Drug delivery systems can also describe the way that drugs are ‘packaged’—like a micelle or a nanoparticle—that protects the drug from degradation and allows it to travel wherever it needs to go in the body. The field of drug delivery has advanced dramatically in the past few decades, and even greater innovations are anticipated in the coming years. Biomedical engineers have contributed substantially to our understanding of the physiological barriers to efficient drug delivery and have also contributed to the development of several new modes of drug delivery that have entered clinical practice.

about images

View More

3D bioprinting

3D bioprinting is an upcoming technique to fabricate tissues and organs through periodic arrangement of various biological materials, including biochemicals and biocells, in a precisely controlled manner. 3D bioprinting has gained momentum in the generation of 3D functional human constructs mimicking native tissues/organs. Various biomaterials based on carbohydrates, proteins, and nucleic acids along with nanocomposites are being used to develop biocompatible and biodegradable scaffolds promoting cell adhesion and proliferation in the tissues engineered using 3D printing. Apart from being biocompatible, polysaccharides are economical and renewable and thus are being explored for the formation of bioinks in regenerative medicine. Extracellular matrix proteins and other structural proteins are known for their impact on tissue mechanics as well as cell behaviour. They have the capability to empower scaffold fabrication with high mechanical strength and porosity and provide cellular mimicry. DNA-based hydrogels and scaffold materials with self-assembling and hybridization properties are ideal for 3D bioprinting. This chapter deals with the principles and applications of 3D bioprinting approach employing the complex biopolymers and nanomaterials and their composites involved in fabrication/regeneration of tissues and organs.

about images
View More