Click
Drag
NTHRYS WHATSAPP
NTHRYS LOGO Welcome Interface Environment Research Products Academics About Contact
NTHRYS LOGO

Plant Pathology Training

Info
Testimonials
FAQs
Placements
Updates

Plant Pathology Training Program

NTHRYS Biotech Labs offers Plant Pathology Training Program under below mentioned protocols. Candidates can opt their interested protocols from the list below. Please click Join button to pay the fee for selected protocol. Fees should be paid individually for all the selected protocols separately by clicking the button. Please save the payment proofs and send them as an attachment to
trainings [ a t ] nthrys [d 0 t] com to receive payment invoices and slot confirmations.
Please Check Modules as well as individual protocols (if any) under this training program. Module has its fee given in the fee structure table and individual fee in its block. Please communicate with our Help Desk Team via whatsapp on +91-9014935156 for any queries.

Modules

Module - I: Detection and Diagnosis

  1. Conventional PCR:
    • DNA Extraction: Isolating DNA from plant tissues using CTAB or commercial kits.
    • DNA Quantification: Estimating DNA concentration using spectrophotometry or fluorometry.
    • Primer Designing: Creating specific primers targeting the pathogen s DNA sequence using software tools like Primer3.
  2. Quantitative Real-Time PCR (qPCR):
    • cDNA Synthesis: Converting RNA to cDNA using reverse transcriptase for RNA targets.
    • Probe Design: Developing fluorescent probes specific to the target DNA sequence.
    • Standard Curve Preparation: For quantification, preparing a standard curve using known concentrations of target DNA.
  3. Reverse Transcription PCR (RT-PCR):
    • RNA Extraction: Extracting RNA from plant tissues using TRIzol reagent or similar kits.
    • cDNA Synthesis: Using reverse transcriptase to convert RNA into cDNA.
    • RNA Quality Assessment: Verifying the integrity and purity of extracted RNA via gel electrophoresis or spectrophotometry.
  4. Loop-mediated Isothermal Amplification (LAMP):
    • DNA Extraction: Similar to PCR, extracting DNA from samples.
    • Primer Set Design: Designing two sets of primers (inner and outer) specific to the target DNA regions.
    • LAMP Reaction Setup: Preparing the reaction mixture with Bst DNA polymerase, primers, and target DNA.
  5. ELISA (Enzyme-Linked Immunosorbent Assay):
    • Antigen Extraction: Extracting plant sap or tissues where the pathogen s antigens are present.
    • Antibody Labeling: Labeling detection antibodies with an enzyme to produce a colorimetric readout upon substrate addition.
    • Plate Coating: Coating microtiter plates with capture antibodies specific to the pathogen antigen.
  6. Immunofluorescence Microscopy:
    • Sample Fixation: Fixing plant tissue samples to preserve structure and antigens.
    • Antibody Incubation: Incubating samples with fluorescently labeled antibodies against the pathogen.
    • Mounting: Preparing samples on slides with a mounting medium for fluorescence microscopy.
  7. Fluorescent in situ Hybridization (FISH):
    • Probe Labeling: Labeling DNA or RNA probes with fluorescent dyes.
    • Hybridization: Denaturing the plant DNA and incubating with labeled probes to allow hybridization.
    • Washing: Removing excess probes to reduce background signal.
  8. Next-Generation Sequencing (NGS):
    • Library Preparation: Preparing DNA/RNA libraries including fragmentation, end repair, and adapter ligation.
    • Sequencing Run: Performing the sequencing using platforms like Illumina or Ion Torrent.
    • Data Analysis: Processing sequencing data for pathogen identification using bioinformatics tools.
  9. Metagenomic Analysis:
    • DNA Extraction: Extracting total DNA from plant samples.
    • Metagenomic Library Preparation: Similar to NGS, but with the aim of capturing a wide array of microbial DNA.
    • Bioinformatics Analysis: Analyzing metagenomic data to characterize the microbial community.
  10. Mass Spectrometry-based Proteomics:
    • Protein Extraction: Isolating proteins from plant tissues.
    • Protein Digestion: Digesting proteins into peptides using enzymes like trypsin.
    • LC-MS/MS Analysis: Separating peptides by liquid chromatography and analyzing them by tandem mass spectrometry.

Module - II: Genetic and Genomic Analysis

  1. Whole Genome Sequencing:
    • Sample Preparation: Isolating high-quality DNA from the pathogen or plant.
    • Library Construction: Preparing sequencing libraries, including shearing DNA, size selection, and adapter ligation.
    • Sequencing: Utilizing platforms like Illumina or Nanopore for high-throughput sequencing.
    • Data Analysis: Employing bioinformatics tools for sequence assembly, annotation, and comparative genomics.
  2. RNA-Seq (RNA Sequencing):
    • RNA Extraction: Isolating total RNA from infected plant tissues.
    • cDNA Synthesis: Converting RNA into cDNA using reverse transcriptase.
    • Library Preparation: Generating RNA-Seq libraries by fragmenting cDNA and adding sequencing adapters.
    • Sequencing and Analysis: Sequencing cDNA fragments and analyzing gene expression data.
  3. sRNA Sequencing:
    • sRNA Isolation: Extracting small RNAs using size selection techniques.
    • Library Preparation: Constructing sRNA libraries with adaptors specific to small RNAs.
    • Sequencing: High-throughput sequencing of small RNA libraries.
    • Data Analysis: Identifying and quantifying small RNAs and their targets using bioinformatics tools.
  4. CRISPR-Cas9 Genome Editing:
    • Target Identification: Selecting gene targets for editing based on disease resistance studies.
    • Guide RNA Design: Designing gRNAs specific to the target gene sequences.
    • Vector Construction: Cloning gRNAs and Cas9 into vectors suitable for plant transformation.
    • Plant Transformation and Screening: Transforming plants with CRISPR constructs and identifying successful edits.
  5. GWAS (Genome-Wide Association Studies):
    • Phenotyping: Accurately measuring disease resistance traits in a plant population.
    • Genotyping: Using SNP arrays or sequencing to genotype the population.
    • Statistical Analysis: Associating genetic variants with phenotypic traits to identify resistance loci.
    • Validation: Confirming the role of identified loci in disease resistance through further genetic studies.
  6. Comparative Genomics:
    • Genome Sequencing: Sequencing genomes of multiple pathogen strains or plant varieties.
    • Alignment and Analysis: Aligning genomes to identify conserved and variable regions.
    • Phylogenetic Studies: Constructing phylogenetic trees to infer evolutionary relationships.
    • Pathogenicity Gene Identification: Identifying genes associated with virulence and host specificity.
  7. Transcriptome Profiling under Stress:
    • Stress Treatment: Exposing plants to pathogens or abiotic stress conditions.
    • RNA Extraction and Sequencing: Isolating RNA from stressed and control plants for sequencing.
    • Differential Expression Analysis: Identifying genes that are upregulated or downregulated in response to stress.
    • Functional Annotation: Annotating the roles of differentially expressed genes in stress response.
  8. Metabolomic Profiling:
    • Metabolite Extraction: Extracting metabolites from plant tissues using solvent extraction methods.
    • LC-MS/MS Analysis: Separating and identifying metabolites using liquid chromatography-mass spectrometry.
    • Data Processing: Analyzing metabolomic data to identify changes in metabolite profiles.
    • Pathway Mapping: Mapping metabolites to biochemical pathways to understand their roles in disease response.
  9. Protein Interaction Mapping:
    • Protein Extraction: Isolating proteins from plant tissues or pathogen cultures.
    • Yeast Two-Hybrid Screening: Identifying protein-protein interactions relevant to disease resistance or pathogenicity.
    • Co-immunoprecipitation (Co-IP): Confirming protein interactions identified by yeast two-hybrid.
    • Mass Spectrometry: Analyzing protein complexes to identify component proteins.
  10. Epigenetic Modifications Analysis:
    • DNA Methylation Analysis: Using bisulfite sequencing to study changes in DNA methylation patterns associated with disease.
    • Chromatin Immunoprecipitation (ChIP): Studying histone modifications and chromatin accessibility changes in response to pathogen attack.
    • ATAC-Seq: Assessing genome-wide chromatin accessibility to identify regulatory elements involved in disease response.
    • RNA Immunoprecipitation (RIP): Investigating interactions between RNA molecules and proteins to study post-transcriptional regulation.

Module - III: Epigenetic and Environmental Interactions

  1. Bisulfite Sequencing:
    • DNA Extraction: Isolating genomic DNA from plant samples.
    • Bisulfite Treatment: Converting unmethylated cytosines to uracils while leaving methylated cytosines unchanged.
    • PCR Amplification: Amplifying the treated DNA to prepare for sequencing.
    • Sequencing and Methylation Analysis: Determining methylation patterns across the genome.
  2. ChIP-Seq (Chromatin Immunoprecipitation Sequencing):
    • Cross-linking: Treating plant cells to cross-link DNA and proteins.
    • Chromatin Shearing: Fragmenting cross-linked chromatin using sonication or enzymatic digestion.
    • Immunoprecipitation: Isolating DNA-protein complexes using antibodies against specific histone modifications or transcription factors.
    • Sequencing: Sequencing the purified DNA to identify regions of the genome associated with the target protein.
  3. ATAC-Seq (Assay for Transposase-Accessible Chromatin using Sequencing):
    • Nuclei Isolation: Extracting nuclei from plant tissues.
    • Transposase Treatment: Tagging accessible chromatin with sequencing adapters using a transposase.
    • PCR Amplification: Amplifying tagged DNA fragments.
    • Sequencing and Analysis: Identifying open chromatin regions across the genome.
  4. RNA Immunoprecipitation (RIP):
    • Cell Lysis and RNA Extraction: Breaking down cells to extract RNA and associated proteins.
    • Immunoprecipitation: Capturing RNA-binding proteins with specific antibodies.
    • RNA Extraction: Isolating RNA from RNA-protein complexes.
    • RT-PCR or Sequencing: Analyzing the associated RNA to identify targets of RNA-binding proteins.
  5. Environmental RNAi:
    • dsRNA Synthesis: Creating double-stranded RNA molecules targeting specific genes of interest.
    • Plant Treatment: Delivering dsRNA to plants via spraying, root absorption, or transgenic expression.
    • Monitoring Gene Silencing: Assessing the knockdown of target gene expression in treated plants.
    • Pathogen Challenge: Evaluating the disease resistance of RNAi-treated plants.
  6. MeDIP-Seq (Methylated DNA Immunoprecipitation Sequencing):
    • DNA Extraction and Fragmentation: Isolating and shearing genomic DNA.
    • Methyl-DNA Immunoprecipitation: Enriching methylated DNA fragments using antibodies against 5-methylcytosine.
    • Library Preparation: Preparing sequencing libraries from enriched DNA.
    • Sequencing and Methylation Profiling: Identifying genome-wide DNA methylation patterns.
  7. Chromatin Conformation Capture (3C) and Hi-C:
    • Cross-linking DNA and Proteins: Stabilizing chromatin structure with formaldehyde treatment.
    • Chromatin Digestion and Ligation: Cutting chromatin with restriction enzymes and ligating proximal DNA ends.
    • De-crosslinking and DNA Purification: Reversing cross-links to purify the DNA.
    • Sequencing: Sequencing ligated DNA fragments to analyze 3D chromatin organization.
  8. Environmental Stress Profiling:
    • Stress Treatment: Exposing plants to various abiotic stresses like drought, salinity, or heat.
    • RNA Extraction and Sequencing: Analyzing changes in gene expression in response to stress.
    • Metabolomic Analysis: Profiling changes in metabolite concentrations under stress conditions.
    • Physiological Measurements: Assessing plant growth, photosynthesis rates, and other physiological parameters in response to stress.
  9. Microbiome Analysis by 16S rRNA Sequencing:
    • Microbial DNA Extraction: Isolating DNA from plant rhizosphere or phyllosphere samples.
    • PCR Amplification of 16S rRNA Genes: Targeting the V4 region or other hypervariable regions specific to bacteria.
    • Library Preparation and Sequencing: Creating sequencing libraries and performing high-throughput sequencing.
    • Data Analysis: Identifying microbial taxa and assessing community structure using bioinformatics tools.
  10. Phenotyping for Epigenetic Variants (Epialleles):
    • Plant Growth and Treatment: Growing plants under controlled conditions and applying treatments that may induce epigenetic changes.
    • High-Throughput Phenotyping: Using imaging and sensor technologies to measure plant traits.
    • Epigenetic Marker Identification: Using bisulfite sequencing or MeDIP-seq to identify epigenetic changes correlated with phenotypic traits.
    • Statistical Analysis: Associating epigenetic markers with observed phenotypes to identify epialleles.

Module - IV: Proteomics, Metabolomics, and Chemical Analysis

  1. Proteomic Analysis by Mass Spectrometry:
    • Protein Extraction: Extracting proteins from plant tissues or cells.
    • Protein Digestion: Treating proteins with trypsin or other proteases to generate peptides.
    • LC-MS/MS Analysis: Separating peptides by liquid chromatography and analyzing by tandem mass spectrometry.
    • Data Analysis: Identifying and quantifying proteins using bioinformatics tools.
  2. Metabolomic Profiling by GC-MS:
    • Metabolite Extraction: Extracting small molecules from plant samples using solvents.
    • Derivatization: Chemically modifying metabolites to make them volatile and amenable to GC-MS analysis.
    • GC-MS Analysis: Separating and identifying metabolites using gas chromatography-mass spectrometry.
    • Data Processing: Analyzing metabolomic data to identify metabolic pathways affected by disease.
  3. Targeted Metabolite Analysis by HPLC:
    • Sample Preparation: Preparing plant extracts for analysis.
    • HPLC Setup: Setting up high-performance liquid chromatography with appropriate columns and detectors.
    • Quantification: Quantifying specific metabolites of interest, such as phytohormones or secondary metabolites.
    • Data Interpretation: Interpreting chromatograms to understand metabolite dynamics in response to pathogens.
  4. Enzyme Activity Assays:
    • Enzyme Extraction: Isolating enzymes from plant tissues.
    • Substrate Incubation: Incubating enzymes with their specific substrates under controlled conditions.
    • Product Quantification: Measuring the amount of product formed using spectrophotometry or fluorescence.
    • Activity Analysis: Calculating enzyme activity levels and comparing them across different treatments or conditions.
  5. Lipidomics by LC-MS:
    • Lipid Extraction: Extracting lipids from plant samples using a mixture of organic solvents.
    • Lipid Separation: Separating lipids by liquid chromatography.
    • Mass Spectrometry: Identifying and quantifying lipids using mass spectrometry.
    • Data Analysis: Interpreting lipidomic data to identify changes in lipid profiles associated with plant diseases.
  6. Phytohormone Analysis by LC-MS/MS:
    • Phytohormone Extraction: Isolating phytohormones from plant tissues using solvent extraction.
    • Sample Preparation: Preparing extracts for LC-MS/MS analysis, including purification and concentration steps.
    • LC-MS/MS Analysis: Quantifying phytohormones using tandem mass spectrometry with specific ionization techniques.
    • Data Interpretation: Analyzing changes in phytohormone levels in response to pathogen infection.
  7. Nuclear Magnetic Resonance (NMR) Spectroscopy for Metabolites:
    • Sample Preparation: Preparing plant extracts for NMR analysis.
    • NMR Spectroscopy: Acquiring NMR spectra to identify and quantify metabolites present in the sample.
    • Data Processing: Using software tools to analyze NMR data and identify metabolite structures.
    • Metabolic Profiling: Profiling the comprehensive metabolite composition of plant samples under various conditions.
  8. Chemical Profiling of Secondary Metabolites:
    • Extraction and Purification: Using solvents and chromatography to extract and purify secondary metabolites from plant tissues.
    • Characterization: Utilizing techniques such as LC-MS, GC-MS, and NMR to characterize the chemical structures of secondary metabolites.
    • Quantification: Determining the concentration of secondary metabolites using calibration curves and analytical techniques.
    • Functional Studies: Investigating the role of specific secondary metabolites in plant defense mechanisms.
  9. Antimicrobial Activity Assays:
    • Microorganism Culturing: Growing pathogenic bacteria or fungi for testing.
    • Compound Application: Applying plant extracts or isolated compounds to microbial cultures.
    • Inhibition Zone Measurement: Measuring the area of growth inhibition around the applied compound on agar plates.
    • Activity Analysis: Analyzing the antimicrobial activity of plant compounds against various pathogens.
  10. Fungicide Resistance Monitoring:
    • Pathogen Isolation: Isolating pathogens from infected plant tissues.
    • Fungicide Application: Exposing isolated pathogens to various concentrations of fungicides.
    • Growth Assessment: Measuring pathogen growth in the presence of fungicides to assess resistance levels.
    • Molecular Analysis: Using PCR and sequencing to identify genetic mutations associated with fungicide resistance.

Module - V: Functional Genomics and Pathogen Interaction

  1. Virus-Induced Gene Silencing (VIGS):
    • Vector Construction: Cloning a fragment of the target gene into a VIGS vector.
    • Agrobacterium Transformation: Transforming Agrobacterium tumefaciens with the VIGS construct.
    • Plant Inoculation: Infiltrating plant leaves with the Agrobacterium carrying the VIGS construct.
    • Phenotypic Observation: Monitoring for silencing effects and disease resistance phenotypes.
  2. Yeast Two-Hybrid (Y2H) Screening:
    • Bait and Prey Construction: Cloning target genes into bait and prey vectors.
    • Yeast Transformation: Introducing bait and prey constructs into yeast strains.
    • Interaction Screening: Growing yeast on selective media to identify protein-protein interactions.
    • Validation: Confirming interactions with additional assays like co-immunoprecipitation.
  3. Co-immunoprecipitation (Co-IP):
    • Protein Extraction: Isolating proteins from plant tissues or cells expressing both bait and prey proteins.
    • Antibody Binding: Incubating the protein mixture with antibodies against the bait protein.
    • Precipitation and Washing: Capturing the antibody-protein complex and washing to remove non-specific binders.
    • Western Blot: Detecting the prey protein in the precipitated complex to confirm interaction.
  4. Bimolecular Fluorescence Complementation (BiFC):
    • Vector Construction: Splitting a fluorescent protein into two halves and fusing each to a protein of interest.
    • Agrobacterium Transformation: Transforming Agrobacterium with BiFC constructs.
    • Plant Transient Expression: Infiltrating plant leaves with Agrobacterium to express the fusion proteins.
    • Fluorescence Microscopy: Observing the reconstituted fluorescence signal in cells where the proteins interact.
  5. RNA Interference (RNAi) for Gene Knockdown:
    • dsRNA Synthesis: Designing and synthesizing double-stranded RNA corresponding to the target gene.
    • Plant Transformation: Delivering dsRNA into plants through Agrobacterium or particle bombardment.
    • Gene Expression Analysis: Quantifying the knockdown of target gene expression by qPCR.
    • Disease Challenge: Assessing the altered susceptibility or resistance to pathogens.
  6. CRISPR-Cas9 for Gene Knockout:
    • Guide RNA Design: Designing guide RNAs targeting specific loci in the plant genome.
    • Vector Construction: Cloning guide RNAs into vectors expressing Cas9 nuclease.
    • Plant Transformation: Introducing the CRISPR construct into plants via Agrobacterium-mediated transformation or biolistics.
    • Genotypic and Phenotypic Analysis: Screening for mutations at the target site and evaluating disease resistance outcomes.
  7. Transient Expression Assays:
    • Construct Preparation: Cloning genes of interest into expression vectors.
    • Agrobacterium Infiltration: Introducing the constructs into plant leaves via Agrobacterium infiltration.
    • Protein Expression Analysis: Verifying the expression of the introduced genes by Western blot or ELISA.
    • Pathogen Challenge: Testing for changes in plant defense response post-transient expression.
  8. Pathogen Host Range Testing:
    • Pathogen Isolation: Culturing different strains or species of pathogens.
    • Inoculation on Different Hosts: Inoculating a range of plant species or varieties with each pathogen.
    • Disease Evaluation: Assessing symptom development and disease progression on different hosts.
    • Molecular Analysis: Using PCR or ELISA to confirm the presence of the pathogen in symptomatic tissues.
  9. Site-Directed Mutagenesis for Protein Function Analysis:
    • Primer Design: Designing primers for mutagenesis to introduce specific mutations into the DNA sequence of the protein of interest.
    • PCR Amplification: Amplifying the target gene with the mutagenic primers.
    • Clone Selection: Selecting and verifying mutated clones through sequencing.
    • Functional Assays: Assessing the impact of mutations on protein function through enzymatic assays or phenotypic analysis in plants.
  10. Protein Localization Studies:
    • Fusion Protein Construction: Fusing the gene of interest with a reporter gene (e.g., GFP).
    • Transient Expression: Introducing the fusion construct into plant cells via Agrobacterium infiltration or particle bombardment.
    • Confocal Microscopy: Observing the subcellular localization of the fusion protein.
    • Co-localization Studies: Introducing markers for specific organelles to study co-localization with the protein of interest.
  11. Pathogen Effector Function Characterization:
    • Effector Gene Cloning: Isolating and cloning effector genes from pathogens.
    • Plant Infiltration: Expressing effector genes in plants using Agrobacterium-mediated infiltration.
    • Phenotype Monitoring: Observing for disease symptoms or defense responses in infiltrated plants.
    • Interaction Studies: Identifying plant targets of pathogen effectors through Y2H screens or co-immunoprecipitation.
  12. Chemical Induction of Plant Defenses:
    • Chemical Treatment: Applying elicitors or defense-inducing chemicals to plants.
    • Gene Expression Analysis: Monitoring changes in expression of defense-related genes post-treatment using qPCR or RNA-Seq.
    • Metabolite Profiling: Analyzing changes in secondary metabolite production using LC-MS or GC-MS.
    • Disease Challenge: Assessing enhanced resistance to pathogens following chemical treatment.
  13. Dual RNA-Seq for Host-Pathogen Interaction:
    • Co-extraction: Isolating both plant and pathogen RNA from infected tissues.
    • Library Preparation and Sequencing: Preparing and sequencing libraries that include both host and pathogen RNA.
    • Data Analysis: Separating and analyzing host and pathogen transcriptomes to study interaction dynamics.
    • Functional Interpretation: Identifying key regulatory genes and pathways involved in the interaction.

Module - VI: Computational Biology and Systems Analysis

  1. Genome Assembly and Annotation:
    • High-throughput Sequencing: Generating raw sequencing data from plant or pathogen samples.
    • Assembly: Using computational tools to assemble the sequencing reads into complete genomes.
    • Annotation: Identifying gene locations and functions within the assembled genomes using bioinformatics software.
    • Database Submission: Uploading annotated sequences to public databases for community access.
  2. Phylogenetic Analysis:
    • Sequence Alignment: Aligning DNA or protein sequences from multiple species or strains.
    • Tree Construction: Employing algorithms to construct phylogenetic trees based on sequence similarities and differences.
    • Evolutionary Inference: Interpreting phylogenetic trees to infer evolutionary relationships and histories.
    • Software Utilization: Using software like MEGA, PhyML, or BEAST for analysis.
  3. Pathway Analysis:
    • Data Gathering: Collecting gene expression data or metabolomic profiles from experiments.
    • Pathway Mapping: Using databases like KEGG to map genes or metabolites to biological pathways.
    • Enrichment Analysis: Identifying significantly impacted pathways using statistical methods.
    • Visualization: Creating diagrams to visualize affected pathways and their components.
  4. Transcriptome Analysis (RNA-Seq Data):
    • Quality Control: Assessing the quality of RNA-Seq reads using tools like FastQC.
    • Read Mapping: Aligning reads to a reference genome or transcriptome with software like STAR or HISAT2.
    • Expression Quantification: Calculating gene expression levels using tools like Cufflinks or HTSeq.
    • Differential Expression Analysis: Comparing expression levels across conditions using DESeq2 or edgeR.
  5. Protein-Protein Interaction Networks:
    • Interaction Data Collection: Compiling known and predicted protein-protein interactions from databases and literature.
    • Network Construction: Using software like Cytoscape to visualize interaction networks.
    • Topological Analysis: Analyzing network properties such as connectivity and centrality measures.
    • Functional Module Identification: Detecting clusters within networks that may represent functional complexes.
  6. SNP and Variant Analysis:
    • Variant Calling: Identifying SNPs and other genetic variants from sequencing data using tools like GATK or Samtools.
    • Annotation: Predicting the effects of variants on genes and proteins using ANNOVAR or SnpEff.
    • Association Studies: Correlating genetic variants with phenotypic traits or disease resistance.
    • Population Genetics Analysis: Examining the distribution and frequency of variants within and across populations.
  7. Metagenomic Analysis:
    • Sequencing Data Processing: Applying quality control and assembly to metagenomic sequencing reads.
    • Microbial Community Profiling: Using tools like QIIME or Mothur to classify microbes and assess community structure.
    • Functional Prediction: Predicting the metabolic capabilities of microbial communities from metagenomic data.
    • Comparative Metagenomics: Comparing microbial communities across different samples or conditions.
  8. Single-Cell RNA-Seq Analysis:
    • Single-Cell Library Preparation: Isolating individual cells and preparing sequencing libraries.
    • Clustering and Cell Type Identification: Analyzing expression data to identify distinct cell populations.
    • Marker Gene Identification: Finding genes that define cell types or states.
    • Pseudotime Analysis: Inferring the developmental trajectory of cells based on gene expression changes.
  9. Machine Learning in Pathogen Prediction:
    • Feature Selection: Identifying relevant features from genomic, transcriptomic, or metabolomic datasets.
    • Model Training: Using machine learning algorithms to build predictive models for disease resistance or pathogen identification.
    • Validation and Testing: Evaluating the model s performance on unseen data to ensure accuracy and reliability.
    • Implementation: Deploying machine learning models for practical applications in pathogen prediction and plant disease management.
  10. Geospatial Analysis for Disease Spread:
    • Data Collection: Gathering geospatial data on disease occurrence and environmental factors.
    • Geospatial Modeling: Using GIS software to model the spatial distribution and spread of plant diseases.
    • Risk Mapping: Creating maps that visualize disease risk levels across different areas.
    • Impact Assessment: Analyzing the potential impact of environmental changes on disease spread and plant health.

Module - VII: Plant Physiology and Stress Responses

  1. Photosynthesis Measurement:
    • Chlorophyll Fluorescence: Using PAM fluorometry to assess the efficiency of photosystem II.
    • Gas Exchange Analysis: Measuring CO2 uptake and transpiration rates using a portable photosynthesis system.
    • Leaf Area and Chlorophyll Content: Estimating photosynthetic capacity through leaf area measurements and chlorophyll extraction.
    • Stress Impact Assessment: Comparing photosynthetic parameters under stress vs. control conditions.
  2. Stomatal Conductance Measurement:
    • Porometer Use: Employing a porometer to measure stomatal conductance directly on leaf surfaces.
    • Water Use Efficiency: Calculating water use efficiency from stomatal conductance and photosynthesis data.
    • Drought Stress Evaluation: Assessing the effect of drought on stomatal behavior and plant water relations.
    • Data Interpretation: Understanding the physiological responses to various stressors through stomatal conductance changes.
  3. Root Growth and Function Analysis:
    • Root Architecture Study: Analyzing root structure and growth patterns using rhizotrons or transparent growth media.
    • Root Exudate Collection: Collecting and analyzing root exudates to study root-microbe interactions.
    • Water and Nutrient Uptake: Measuring uptake rates to assess root function under stress conditions.
    • Mycorrhizal Association Assessment: Evaluating the impact of mycorrhizal fungi on root health and stress resilience.
  4. Plant Hormone Analysis:
    • Hormone Extraction: Isolating hormones from plant tissues using solvent extraction.
    • LC-MS/MS Quantification: Identifying and quantifying phytohormones with liquid chromatography-tandem mass spectrometry.
    • Hormone Profiling: Comparing hormone levels across different treatments or developmental stages.
    • Functional Studies: Correlating hormone levels with physiological responses or stress tolerance.
  5. Osmotic and Salinity Stress Assays:
    • Electrolyte Leakage Measurement: Assessing cell membrane integrity under stress conditions.
    • Osmolyte Quantification: Measuring levels of proline, glycine betaine, and other osmolytes that contribute to osmotic adjustment.
    • Ion Content Analysis: Determining Na+ and K+ concentrations in tissues to assess ion homeostasis under salinity stress.
    • Growth and Yield Analysis: Evaluating the impact of osmotic and salinity stress on plant growth and productivity.
  6. Heat and Cold Stress Response Evaluation:
    • Thermotolerance Assays: Assessing plant survival and recovery after exposure to high or low temperature shocks.
    • Heat Shock Protein (HSP) Analysis: Measuring the expression of HSPs as indicators of heat stress response.
    • Chilling Injury Assessment: Evaluating symptoms and physiological markers of cold stress in plants.
    • Molecular Markers of Cold Acclimation: Identifying gene expression changes associated with cold tolerance.
  7. Heavy Metal Stress Analysis:
    • Heavy Metal Accumulation: Measuring metal concentrations in plant tissues using atomic absorption spectroscopy or ICP-MS.
    • Antioxidant Enzyme Activity: Assessing the activity of enzymes like superoxide dismutase and catalase in response to metal stress.
    • Phytoremediation Potential: Evaluating the ability of plants to tolerate and accumulate heavy metals from contaminated soils.
    • Gene Expression Studies: Analyzing the expression of metal transporters and chelators involved in metal detoxification.
  8. Reactive Oxygen Species (ROS) Detection:
    • ROS Staining: Using fluorescent dyes like DCFH-DA to visualize ROS accumulation in plant tissues.
    • Enzyme Activity Assays: Measuring activities of ROS-scavenging enzymes such as catalase and peroxidase.
    • Lipid Peroxidation Measurement: Estimating cell membrane damage through malondialdehyde (MDA) content.
    • Antioxidant Capacity: Assessing the total antioxidant capacity of plant extracts using assays like FRAP or DPPH.
  9. Abiotic Stress Tolerance Screening:
    • Germination Tests: Evaluating seed germination rates under various abiotic stress conditions.
    • Stress Tolerance Indices: Calculating indices based on growth, yield, and physiological parameters under stress vs. control conditions.
    • Molecular Breeding: Using markers associated with stress tolerance for breeding more resilient plant varieties.
    • Transgenic Approaches: Developing and testing transgenic lines with enhanced stress tolerance traits.
  10. Signal Transduction Pathway Elucidation:
    • Pathway Inhibitor Studies: Using specific chemical inhibitors to dissect signaling pathways involved in stress responses.
    • Reporter Gene Assays: Employing transgenic plants with reporter genes under the control of stress-responsive promoters to monitor signaling activity.
    • Protein Kinase Activity: Measuring the activity of kinases involved in signal transduction during stress.
    • Calcium Signaling Studies: Investigating the role of calcium as a second messenger in stress-induced signaling pathways.

Module - VIII: Ecological and Evolutionary Dynamics

  1. Plant-Pathogen Co-evolution Studies:
    • Comparative Genomics: Analyzing the genomes of plants and their pathogens to identify co-evolutionary patterns.
    • Cross-Infection Experiments: Testing pathogen infectivity across a range of host species to study adaptation and host specificity.
    • Molecular Evolution Analysis: Assessing the evolutionary rates and selection pressures on genes involved in host-pathogen interactions.
    • Phylogenetic Reconstruction: Constructing phylogenies to elucidate the co-divergence between pathogens and their hosts.
  2. Microbial Community Dynamics in Rhizosphere:
    • Soil DNA Extraction: Isolating microbial DNA from the rhizosphere for metagenomic analysis.
    • 16S rRNA Sequencing: Profiling bacterial communities to understand the composition and diversity of the rhizosphere microbiome.
    • Functional Metagenomics: Sequencing the total DNA to predict functional capabilities of the rhizosphere microbiome.
    • Plant Growth Promotion Assays: Testing the effect of specific microbial communities on plant growth and health.
  3. Soil Health and Plant Disease Suppression:
    • Soil Physicochemical Analysis: Measuring soil properties like pH, organic matter content, and nutrient levels.
    • Suppressiveness Assays: Evaluating soil s ability to suppress plant diseases through bioassays.
    • Microbial Inoculation Studies: Investigating the effect of adding beneficial microbes to soil on disease suppression.
    • Molecular Markers for Soil Health: Developing markers to assess the biological quality and health of soil.
  4. Invasive Species and Pathogen Spread:
    • Geographic Mapping: Tracking the spread of invasive species and pathogens using GIS tools.
    • Population Genetics: Analyzing genetic diversity and population structure of invasive species to understand their spread.
    • Biological Control Strategies: Assessing the effectiveness of biological control agents against invasive species.
    • Policy and Management: Developing strategies and policies for managing invasive species and preventing further spread.
  5. Climate Change Effects on Plant Diseases:
    • Climate Modeling: Predicting changes in disease distribution and severity under future climate scenarios.
    • Phenology Studies: Observing changes in plant and pathogen life cycles in response to climate variables.
    • Host Range and Virulence: Studying how changing climates affect pathogen host range and virulence.
    • Adaptation Strategies: Developing plant breeding and management strategies to cope with climate-induced disease challenges.
  6. Biodiversity and Ecosystem Services in Agriculture:
    • Agroecosystem Analysis: Evaluating the role of biodiversity in supporting ecosystem services like pollination and pest control.
    • Conservation Agriculture Practices: Implementing farming practices that enhance biodiversity and ecosystem services.
    • Ecosystem Service Valuation: Assessing the economic value of ecosystem services provided by biodiversity in agricultural landscapes.
    • Policy and Conservation Measures: Developing policies to protect and enhance biodiversity in agricultural ecosystems.
  7. Herbivore-Plant Interactions:
    • Herbivory Impact Studies: Measuring the effects of herbivore damage on plant growth, reproduction, and susceptibility to pathogens.
    • Induced Defense Mechanisms: Investigating plant responses to herbivory, such as the production of defensive chemicals.
    • Trophic Interactions: Understanding the interactions between plants, herbivores, and predators in the context of plant disease dynamics.
    • Chemical Ecology: Studying the chemical signals involved in plant-herbivore and herbivore-predator interactions.
  8. Endophytic and Epiphytic Microbes in Plant Health:
    • Isolation and Characterization: Identifying endophytic and epiphytic microbes associated with healthy plants.
    • Functional Studies: Assessing the roles of these microbes in enhancing plant growth and disease resistance.
    • Symbiosis Mechanisms: Elucidating the molecular and biochemical mechanisms underlying plant-microbe symbioses.
    • Application in Agriculture: Exploring the use of beneficial microbes as biofertilizers or biopesticides.
  9. Genetic Resources for Disease Resistance:
    • Gene Bank Utilization: Accessing genetic resources from gene banks for breeding disease-resistant varieties.
    • Wild Relatives: Exploring the disease resistance traits of wild relatives of crop plants.
    • Molecular Breeding: Incorporating resistance genes from diverse genetic sources into cultivars using marker-assisted selection.
    • Conservation Strategies: Developing strategies for the conservation of genetic diversity important for disease resistance.
  10. Integrated Disease Management:
    • Cultural Practices: Implementing crop rotation, intercropping, and other practices to reduce disease incidence.
    • Biological Control: Utilizing beneficial microbes or natural enemies to manage plant pathogens.
    • Chemical Control: Applying fungicides or other chemical controls judiciously as part of an integrated disease management strategy.
    • Resistance Breeding: Developing and deploying disease-resistant plant varieties as a foundational component of disease management.

Please contact us via WhatsApp for fee details on +91-9014935156

Please choose a suitable time slot and inform our team via WhatsApp on +91-9014935156 (located at the top right corner) to receive the payment link for fee payment and slot confirmation.

Training based on Individual Protocols



DNA Extraction from Plant Leaf
Fee: ₹ 1680/-
Time: 6 Hours
Join
Optimization of PCR parameters - Technical Theory - -No practical
Fee: ₹ 360/-
Time: 1 Hours
Join
A brief introduction to Plant Bioinformatics -Nomenclature and Plant Pathological Bioinfo Database designing and management standards
Fee: ₹ 1800/-
Time: 3 Hours
Join
DNA extraction from Fungal Plant Pathogens
Fee: ₹ 3600/-
Time: 5 Hours
Join
DNA Extraction from Viral Plant Pathogens
Fee: ₹ 6000/-
Time: 8 Hours
Join
Identification of Fungal Plant Pathogens using PCR
Fee: ₹ 7200/-
Time: 12 Hours
Join
Identification of Viral Plant Pathogens using PCR
Fee: ₹ 10800/-
Time: 12 Hours
Join
DNA extraction from Insect Plant Pathogens
Fee: ₹ 8400/-
Time: 8 Hours
Join
Identification of Insect Plant Pathogens using PCR
Fee: ₹ 10800/-
Time: 12 Hours
Join
Total Protein Extraction from plant materials
Fee: ₹ 18000/-
Time: 8 Hours
Join
Semi Ultra Purification of extracted plant proteins
Fee: ₹ 8400/-
Time: 6 Hours
Join
Immunological Identification of plant pathogens using ELISA
Fee: ₹ 4800/-
Time: 8 Hours
Join
Microbiological Quality Assurance Measures in Plant Tissue Culture Practices
Fee: ₹ 12000/-
Time: 1 Hours
Join
Stock Plant treatment for detection and Identification of viriods viruses bacteria and fungi in plant tissue culture plant materials
Fee: ₹ 18000/-
Time: 8 Hours
Join
Various Surface sterilization to control microbial hazards plant tissue culture plant materials
Fee: ₹ 600/-
Time: 1 Hours
Join
Molecular identification of Viral contamination of plant material selected for plant tissue culture
Fee: ₹ 30000/-
Time: 8 Hours
Join
Identification of Plant Disease Resistance Genes
Fee: ₹ 14400/-
Time: 8 Hours
Join

Note: NTHRYS currently operates through three registered entities: NTHRYS BIOTECH LABS (NBL), NTHRYS OPC PVT LTD (NOPC), and NTHRYS Project Greenshield (NPGS).

NTHRYS CONSORTIUM

NBL NOPC NPGS
Updates
Industrial

Basic

Academic

Terms | Refund and Cancellations | Privacy Policy | Shipping Policy | Retruns Policy
Website Designed, Programmed and Maintained
by
Balaji S Rao


Web

Chat
Contact

Us
Download

PDF
Placements