Call for Abstract

16th International Conference on Proteomics, Genomics & Bioinformatics, will be organized around the theme “”

PROTEOMICS 2023 is comprised of keynote and speakers sessions on latest cutting edge research designed to offer comprehensive global discussions that address current issues in PROTEOMICS 2023

Submit your abstract to any of the mentioned tracks.

Register now for the conference by choosing an appropriate package suitable to you.

Immunogenetics and Immunology

Immunogenetics, sometimes known as immunogenetics, is a branch of medical genetics that studies the link between genetics and the immune system. Autoimmune disorders, such as type 1 diabetes, are complicated hereditary features caused by immune system malfunctions.

The biological and molecular basis for the body's defence against germs (such as bacteria, viruses, and fungi), as well as outside agents such as biological poisons and environmental pollutants, as well as failures and malfunctions of these defence mechanisms, are studied in immunology. Aside from external influences on the organism, there are also defence reactions involving the body's own cells, such as in the case of bodily reactions to cancer and the absence of a body's reaction to healthy cells in the case of an immune-mediated sickness. As a result, immunology is a branch of biology. Edward Jenner, who discovered in 1796 that cowpox, or vaccinia, induced immunity against human smallpox, is generally credited with its invention.

The word immunogenetics refers to all processes in an organism that are governed and impacted by the organism's genes on the one hand, and are significant in terms of the organism's immunological defence reactions on the other.

 

Proteomics and its Medicinal Research

Instead of treating patients with a "one size fits all" approach, customised drug programmes allow doctors to adapt medications to achieve the greatest results for individual patients. Customized prescription, additionally named exactness pharmaceutical, is a medicinal methodology that isolates patients into various gatherings—with restorative choices, practices, mediations as well as items being custom-made to the individual patient in view of their anticipated reaction or danger of malady.

 

Metabolomics in Precision Medicine

Metabolomics, or the post-genomic investigation of the particles and strategies that make up the absorption framework, looks to be a potentially revolutionary new way of looking at science and disease. Accuracy Medicine is a method of locating and manufacturing medications and antibodies that provides patients with unparalleled outcomes by combining clinical and sub-nuclear data to determine the natural occurrence of contamination. Pharmacometabolomics complements and educates pharmacogenomics, and the two combined provide the foundation for Quantitative and Systems Pharmacology. Accuracy provides cutting-edge solutions to help you achieve more success in translational research. The arrival of massive parallel sequencing reflects a shift in biomarker disclosure and clinical preclinical planning as we move closer to what is now known as "biomarker-driven tumour solution" or "accuracy medication." The President's 2016 Budget will provide a $215 million theory to the National Institutes of Health (NIH), the Food and Drug Administration (FDA), and the Office of the National Coordinator for Health Information Technology (ONC) to support this effort, in addition to generous dares to comprehensively reinforce investigation, change, and progress.

Pharmacometabolomics is a field in which massive biochemical data capturing the effects of the genome, gut microbiota, and condition exposures is being used to find data on metabotypes and treatment outcomes, and metabolic markings are being developed as new potential biomarkers. The Precision Medicine Initiative gives the National Cancer Institute $70 million to research tumour genomic drivers, identify those goals, and develop new treatments. Exactness Medicine refers to tailoring restorative treatment to each patient's unique characteristics. It doesn't necessarily imply the creation of one-of-a-kind medications or therapeutic devices for each patient, but rather the ability to divide people into subpopulations that differ in their susceptibility to a specific illness, in the science or potential visualisation of the ailments they may cause, or in their response to a specific treatment.

 

Metabolic Syndrome

Metabolomics is used to investigate a variety of human diseases, improve their assurance and repugnance, and design better accommodating systems. Novel biomarkers and mechanisms of cardiovascular disease danger have been identified using metabolomic profiling. Sustenance and Metabolism Center and Center for Human Genetics at Duke University where the examination is proceeding with National Institutes of Health grants . Metabolomics can give certain inclinations in regard to other "omics" progressions (genomics, transcriptomics, proteomics) in diabetes look into. CEDAM (Center for Endocrinology, Diabetes and Metabolism) investigation is centered around a seat to-bedside approach, taking examination through from key science divulgence to clinical application. This is energized by recurring pattern MRC Experimental Medicine and Biomarker Grants and enhanced by the close-by region of lab workplaces, Welcome Trust Clinical Research Facility and the Queen Elizabeth Hospital (University Hospital NHS Foundation Trust). Metabolomic approach gives novel encounters into the robotic investigations of antitumor solutions from a point specific from conventional drug examinations. For metabolomics approach change, distinct novel pathways have been used. The use of metabolomics in a variety of distinct examination zones spanning fundamental, biological, and clinical sciences has been pushed forward by recent breakthroughs.

Novel indicators and components of cardiovascular infection risk have been identified using metabolomic profiling. Metabolomics requirements for bioinformatics include information and data management, raw scientific data preparation, metabolomics measures and metaphysics, factual examination and data mining, data incorporation, and numerical demonstrating of metabolic systems within a framework of frameworks science. The real approaches in metabolomics, as well as cutting-edge scientific instruments used in the information age, are examined in relation to these specific bioinformatics requirements. Endocrinology is a therapeutic specialty that deals with the investigation and treatment of hormone-related illnesses, particularly biochemical operations related to the body's normal functioning. Metabolomics allows for a more detailed analysis of metabolites and has been linked to the discovery of biomarkers and irritated pathways that can help clear up the activity component of traditional Chinese medicines (TCM).

 

Urine Metabolomics

Urine metabolomics emerged in response to a specific disease or therapeutic intervention for the discovery of non-invasive biomarkers that can detect small metabolic abnormalities. Urine is distinguished from other biofluids by its ease of collection, abundance of metabolites, and capacity to reflect abnormalities in all metabolic pathways within the body. To quench any biogenic and/or non-biogenic chemical processes, urine samples for metabolomic analysis must be quickly refrigerated. Because of the constant advances in its linked technologies, liquid chromatography (LC)-MS is without a doubt the most analytical technique used in urine metabolomics. The Urine Metabolome database includes detailed information on 3100 small molecule metabolites discovered in human urine, as well as 3900 concentration values. The metabolite data columns have hyperlinks to other databases (KEGG, PubChem, ChEBI, Chemspider, DrugBank, PDB and Uniprot). Chemical, clinical, and molecular/biochemistry data are all used to create this information.

 

Cancer Proteome and Biomarkers

Proteomics technologies are employed in the discovery of novel therapeutic medicines and for early identification and diagnosis of malignancies. With advancements in the science of proteomics and the use of mass spectrometry, breast cancer observation, prognosis, diagnosis, and treatment are now possible.

With the development of proteomics technology, researchers have been able to distinguish between disease and disease-free states linked with breast cancer. Proteins expressed or discovered in serum, plasma, and tumour cells employing novel approaches provide for a better understanding of cancer heterogeneity. Proteomics in Breast Cancer Proteomics in Skin Cancer Proteomics in Lung Cancer Proteomics in Ovarian Cancer Proteomics in Colorectal Cancer.

 

Cardiovascular Proteomics

Proteomics has arrived at a critical juncture in cardiovascular research. Analyses of cardiac and vascular illness at the organ, subcellular, and molecular levels have shown dynamic, complicated, and nuanced intracellular mechanisms. Proteomic analysis' power and flexibility, which make protein separation, identification, and characterisation easier, should speed up our understanding of these processes at the protein level. Proteomics, when used correctly, gives researchers with cellular protein "inventories" at precise points in time, making it excellent for recording protein alteration as a result of a specific disease, condition, or treatment. This is performed by creating species- and tissue-specific protein databases, which serve as a foundation for future proteomic research. The advancement of proteomic techniques has allowed for a more detailed analysis of the molecular pathways behind cardiovascular illness, allowing for the discovery of both changed proteins and the type of their modification. Continued development should lead to functional proteomic studies, in which the identification of protein modification, in combination with functional data from established biochemical and physiological methods, can help us better understand how the proteome changes in response to cardiovascular disease.

 

Quantitative Proteomics

Quantitative Proteomics is a scientific technique for determining the amount of protein in a sample, which may then be used to compare diseased and healthy people. It also provides information on sample differences. Isotopic labelling of proteins or peptides can be differentiated using mass spectrometry. Other life science domains such as genomics, kinemics, transcriptomics, and metabolomics are increasingly using broad-scope analysis, and the quantitative Proteomic approach is in line with that. Quantitative Dot Blot (QDB) Electrospray Ionization 2DE-DIGE Mass Spectroscopy Optimizing LC-Ms/MS for Quantitative Proteomics.

Quantification using spectrophotometry

Spectrophotometric methods can be used to determine the concentration of a specific protein in a sample. A spectrophotometer may be used to measure the OD at 280 nm of a protein, which can then be used in conjunction with a standard curve assay to quantify the content of Tryptophan, Tyrosine, and Phenylalanine. This method, however, is not the most accurate because protein composition varies widely, and this method would be unable to quantify proteins that do not contain the aforementioned amino acids. Due to the likelihood of nucleic acid contamination, this approach is also imprecise. Biuret, Lowry, BCA, and other more accurate spectrophotometric techniques for protein quantification.

 

Protein Interactions in Biology

Because of the inherent complexity of signalling networks, as well as the quantity and variety of quantitative data, cell signalling systems and networks can be examined using system biology. Through cell signalling, a vast spectrum of stimulus-response behaviours is observed in cells, which is important to all of Biology. The primary function of cell signalling systems is to take information from the environment and generate an output response based on that input. These signalling pathways constitute the foundation for many human disorders, and they show great interest in medicine and human health. Physical Interaction networks can define Interacting Protein Pairs according to Physio-Chemical Principles. This type of matrix aids the integration of structural and system biology. Networks of metabolism Co-expression networks of genes Interactions between proteins and DNA Networks of Neurons Interactions between proteins.

 

Proteomics in Biomedical Applications

Proteomics has its origins in two-dimensional gel electrophoresis (2-DE), a technique developed more than twenty years ago. 2-DE has a high-resolution capacity, and was initially used primarily for separating and characterizing proteins in complex mixtures. 2-DE is still a useful method for identifying proteins, but it is now frequently used in conjunction with mass spectrometry (MS), a technique that has evolved significantly in recent years. The recent completion of the human genome project has resulted in a vast DNA database that can be used by bioinformatics, and the next task for scientists is to discover an organism's whole proteome. The integration of genomic and proteomic data will aid in the understanding of protein activities in disease pathogenesis and the ageing process, and may lead to the discovery of novel therapeutic target proteins and disease biomarkers. This study explores the possible applications of proteomics in biological research and describes current breakthroughs in proteomic technology.

 

Gene Therapy

Human gene therapy aims to alter the biological characteristics of living cells or adjust the expression of a gene for therapeutic purposes.

Gene therapy is a strategy for treating or curing disease by altering a person's DNA. Gene treatments can function in a variety of ways:

  • Putting a healthy copy of a disease-causing gene in place of the disease-causing gene
  • Inactivating a disease-causing gene that isn't working as it should
  • To assist treat an illness, a new or modified gene is introduced into the body.

Cancer, hereditary illnesses, and infectious diseases are among the ailments for which gene therapy products are being researched.

 

Epigenomics

Epigenomics is the study of the epigenome, or the whole set of epigenetic alterations on a cell's genetic material. The field is comparable to genomics and proteomics, which examine a cell's genome and proteome. Reversible alterations to a cell's DNA or histones that affect gene expression without changing the DNA sequence are known as epigenetic modifications. Epigenomic maintenance is an ongoing process that contributes to the stability of eukaryotic genomes by participating in critical biological systems such as DNA repair. Plant flavones are thought to inhibit cancer-causing epigenomic markers.  DNA methylation and histone modification are two of the most well-studied epigenetic changes. Epigenetic changes are engaged in a variety of cellular processes, including differentiation/development and cancer, and play a key role in gene expression and regulation. Epigenetics research on a worldwide scale has only lately been possible thanks to the application of genomic high-throughput techniques.

 

Proteomics

Proteomics is the study of proteomes on a vast scale. A proteome is a collection of proteins made by a living creature, system, or biological milieu. We can talk about a species' proteome (for example, Homo sapiens) or an organ's proteome (for example, the liver). The proteome is dynamic, varying from cell to cell and changing throughout time. The proteome reflects the underlying transcriptome to some extent. However, in addition to the expression level of the relevant gene, many other factors influence protein activity (which is generally measured by the response rate of the processes in which the protein is engaged).

Proteomics is used to investigate:

  • when and where proteins are expressed
  • protein synthesis, degradation, and steady-state abundance rates
  • how proteins are modified (for example, phosphorylation and other post-translational modifications (PTMs))
  • protein transport between subcellular compartments
  • proteins' involvement in metabolic pathways
  • what happens when proteins come into contact with one another

 

Pharmacogenomics

The study of the role of the genome in medication response is known as pharmacogenomics. The combination of pharmacology and genetics is reflected in the term (pharmaco- + genomics). Pharmacogenomics studies how a person's genetic composition influences their pharmacological reaction. It examines the impact of acquired and inherited genetic variation on drug response in patients by linking gene expression or single-nucleotide polymorphisms to pharmacokinetics (drug absorption, distribution, metabolism, and elimination) and pharmacodynamics (effects mediated by a drug's biological targets). The terms pharmacogenomics and pharmacogenetics are frequently used interchangeably. Although both names refer to medication response influenced by genetic factors, pharmacogenetics focuses on single drug-gene interactions, whereas pharmacogenomics takes a more genome-wide approach, including genomics and epigenetics while addressing the impact of numerous genes on drug response.

 

Integrating Transcriptomics & Proteomics

Through multiple controlled processes, protein-coding genes are transcribed to pre messenger ribonucleic acid (pre-mRNA), then processed to messenger ribonucleic acid (mRNA), and finally translated into protein. Proteins can be further processed and changed post translationally, and they can form complexes with one another. The transcriptome is the whole set of coding and noncoding RNA molecules, while the proteome is the complete set of proteins expressed in an organelle, cell type, or tissue under specified conditions. Transcriptomics is a set of high-throughput technologies that provides information on the sequencing and quantity of transcripts. Proteomics is a group of techniques that have been developed to provide identification of proteins, their expression levels, and posttranslational changes at a high-throughput level. Increases in transcript levels may not necessarily equate to similar changes in protein levels, and the two disciplines provide complementary information.

 

Protein Sequencing

Protein sequencing is the technique of determining the amino acid sequence of a protein or peptide in its entirety or in part. This could be used to identify the protein or define the post-translational alterations it has undergone. Partially sequencing a protein usually offers enough information (one or more sequence tags) to identify it using databases of protein sequences acquired via conceptual gene translation. Mass spectrometry and Edman degradation with a protein sequenator are the two most common direct methods of protein sequencing (sequencer). Although mass spectrometry is now the most extensively used approach for protein sequencing and identification, Edman degradation is still a useful tool for determining a protein's N-terminus.

 

Machine learning in Bioinformatics

The use of machine learning techniques to bioinformatics, such as genomics, proteomics, microarrays, systems biology, evolution, and text mining, is known as machine learning in bioinformatics. High-performance computing allows important breakthroughs in bioinformatics.

Prior to the invention of machine learning algorithms, bioinformatics programmes had to be explicitly designed by hand, which was incredibly difficult for tasks like protein structure prediction. Automatic feature learning is enabled by machine learning techniques like as deep learning, which allow the algorithm to learn how to combine various aspects of the input data into a more abstract collection of features from which to conduct additional learning based on the dataset alone. When trained on huge datasets, this multi-layered method allows such computers to generate extremely complicated predictions. Other computational biology approaches, on the other hand, while still capable of dealing with enormous datasets, do not allow the data to be interpreted and evaluated only by the engine. The quantity and amount of available biological datasets have exploded in recent years, allowing bioinformatics researchers to apply machine learning algorithms.

 

System Biology

The computer and mathematical analysis and modelling of complex biological systems is known as systems biology. It is a biology-based interdisciplinary branch of study that focuses on complex interactions within biological systems and employs a holistic approach to biological research (holism rather than reductionism).

The term has been widely employed in biology in a number of circumstances, particularly after the year 2000. The Human Genome Project is an example of applied systems thinking in biology that has resulted in new, collaborative ways of working on genetics challenges. One of the goals of systems biology is to model and find emergent properties, which are traits of cells, tissues, and organisms that function as a system and can only be described theoretically using systems biology techniques. Typically, metabolic or cell signalling networks are involved.

Computational Biology

The science of using biological data to construct algorithms or models in order to understand biological systems and relationships is known as computational biology, which encompasses many areas of bioinformatics. Biologists did not have access to enormous amounts of data until recently. This information is now widely available, particularly in the fields of molecular biology and genomics. Researchers were able to establish analytical methods for analysing biological data, but they were unable to immediately communicate them with their peers.

 

 

Structural Genomics

The goal of structural genomics is to describe the three-dimensional structure of each protein encoded by a genome. By combining experimental and modelling methodologies, this genome-based methodology provides for a high-throughput method of structure determination. The main distinction between structural genomics and standard structural prediction is that structural genomics tries to figure out the structure of every protein encoded by the genome rather than just one. Structure prediction can be done more quickly using a combination of experimental and modelling approaches now that full-genome sequences are available, especially because the availability of a large number of sequenced genomes and previously solved protein structures allows scientists to model protein structure on previously solved homologs.

 

Protein Structure

The linear sequence of amino acids in a peptide or protein is known as primary structure. The main structure of a protein is described from the amino-terminal end to the carboxyl-terminal end, as is customary. Ribosomes are the most frequent organelles in cells that perform protein production.

Genomics

The structure, function, evolution, mapping, and editing of genomes are all studied in genomics, which is an interdisciplinary subject of biology. A genome is a full set of DNA that includes all of an organism's genes. In contrast to genetics, which focuses on individual genes and their functions in inheritance, genomics tries to characterise and quantify all of an organism's genes, as well as their interrelationships and effects on the organism as a whole. With the help of enzymes and messenger molecules, genes may direct the production of proteins. Proteins, in turn, are responsible for the formation of body structures such as organs and tissues, as well as the management of chemical reactions and the transmission of information between cells. Genomics also entails the assembly and analysis of complete genomes using high-throughput DNA sequencing and bioinformatics to assemble and study their function and structure. Advances in genomes have sparked a revolution in discovery-based research and systems biology, making even the most complicated biological systems like the brain easier to comprehend.

 

Bioinformatics

Bioinformatics is an interdisciplinary science that develops methods and software tools for analysing biological data, particularly big and complicated data sets. Bioinformatics is an interdisciplinary discipline of research that analyses and interprets biological data by combining biology, computer science, information engineering, mathematics, and statistics. Bioinformatics has been utilised for mathematical and statistical in silico assessments of biological questions.

Bioinformatics include biological research that employ computer programming as part of their approach, as well as a set of commonly used analysis "pipelines," particularly in the field of genomics. Bioinformatics is commonly used to identify candidate genes and single nucleotide polymorphisms (SNPs). Such identification is frequently done in order to better understand the genetic basis of disease, unique adaptations, attractive features (especially in agricultural species), or population variances. Bioinformatics also aims to comprehend the organisational principles within nucleic acid and protein sequences, known as proteomics, in a less formal fashion.