Review Article - (2025) Volume 16, Issue 2
Received: 24-May-2024, Manuscript No. JPPM-24-25842; Editor assigned: 29-May-2024, Pre QC No. JPPM-24-25842 (PQ); Reviewed: 12-Jun-2024, QC No. JPPM-24-25842; Revised: 12-May-2025, Manuscript No. JPPM-24-25842 (R); Published: 19-May-2025, DOI: 10.35248/2157-7471.25.16.753
The approach that is focused on the integration of multiomics data is now the most efficient way of revealing the complexity underlying biological systems and improving the knowledge of health and disease. In this piece, we focus on how and how multiomics data are combined to provide a full picture of biological processes as well as the challenges involved. Data integration, network reconstruction and pathway analysis are some of the computational frameworks that are well covered in this review. By highlighting the diverse approaches used to harmonize omics datasets and untangle biological interactions, omics discovery can be performed at another level. Moreover, we are investigating the utility of multiomics integration in biomarker identification, disease typing, drug discovery and precision medicine and highlight the potential role of multiomics in triggering disruptive discoveries in fundamental science as well as in translation. Finally, we address some basic problems that multiomics integration has and specify how the field may be developed by contributing to standardization, scalability and data sharing. This paper embraces the novelty of multiomics integration and its connotations for precision medicine and individual-targeted healthcare. Overall, we manage to offer readers some knowledge of new fields of multiomics research contributing to precision medicine and pertinent health care.
Fugus; Pathogens; Omics; Maize
Importance of maize and its susceptibility to fungal pathogens: Maize is an important staple food crop in various regions of Pakistan and is an important agricultural asset. It is grown globally for food, biofuel, animal feed ingredients and many other bioproducts. Its importance is that it impacts the economy of the United States annually by US $75 million. It is grown in temperate and tropical regions and hence provides an important livelihood for local farmers. However, farmers face many challenges amidst the high humidity and high temperature in the regions of Sub-Saharan Africa. The potential of different types of fungi to withstand heat and humidity from scorching poses a significant risk to farmers and many ears of maize are lost through mycotoxins produced by these fungal pathogens. Maize has silk, which provides a pathway for fungal spores to germinate and grow alongside maize and affects the maize crop. There are various species that can notably affect maize plants:
•Fusarium verticillioides
•Aspergillus flavus link
•Speare of Aspergillus parasiticus
The potential of these species to grow in preharvest kernels of maize puts it at risk for damage and rotting of the important maize crop, which acts as a nuisance for the farmers growing maize. However, the storage infrastructure also provides a chance for increased spoilage due to inadequate mycotoxin testing and high humidity levels in different regions. The different accidental pathways that can lead to fungal infection include insect cuts and style or thread-like channels called silk. The susceptibility of maize to the fungus increases through this silk part of the maize, which in turn acts as a moisture-filled, soft, nutrient-filled material with protected leaves wrapped inside. These features make it a lovely atmosphere for fungi to temporarily stay there and then make their permanent stay in the seed. However, the silk is the female part of the plant where pollination occurs and maize seeds are grown through double fertilization.
Types of fungal pathogens affecting maize: As we discussed earlier, there are notable fungal pathogens that can affect maize. In fact, the fungus itself is not harmful just as long as it remains in a communalistic relationship. However, some fungal species produce harmful toxins, such as mycotoxins, including DON toxins and aflatoxins, which are toxic to human consumption as well as animal health. F. graminearum is a notable fungus that causes Gibberellic ear rot. This type of fungus produces dangerous levels of DON and zearalenone that can initiate many symptoms in humans, such as vomiting, diarrhea, fever and headaches and estrogen hormonal disturbance. The peculiar nature of this toxin is that it does not degrade at high cooking temperatures. Fusarium ear rot is another type of fungus that produces the harmful carcinogenic pathogen fumonisin, which is the cause of many cancers, including esophageal cancer and liver cancer. In Kenya in 2006, fumonisin growth was reported in 86% of the total samples tested. The germination and pollination phenomena provide support for the development of this type of fungal pathogen. Aspergillus flavus and Aspergillus parasiticus are also fungi that produce dangerous levels of aflatoxins that affect the human population. It directly affects the liver through chronic infection and has mutagenic and teratogenic effects in humans. The other important aspect of this fungal pathogen is that it first invades the livestock population, after which it moves through the food chain hierarchy and poisons humans.
Significance of studying the impact of fungal pathogens on maize growth: The effects of mycotoxins on humans are a greater concern for public health departments, so this study focused on the impact of these pathogens on maize growth. The purpose of this study was to determine a safe parameter to determine how maize crops are affected by different routes of contamination and to devise ways to reduce the possibility of fungal infection. To study the impact of pathogens on maize growth, we must first determine how maize kernels are infected and the environmental factors that cause maize rot. By studying this topic, we will prepare for future crops to avoid damage by inventing new insecticides. Studying the interaction between corn-infecting fungus and corn plants will facilitate the development of scientific knowledge and innovation in the field of plant pathology and to some extent, agricultural biotechnology. Through cross-breeding or genetic engineering, researchers can identify the types of maize that are resistant to diseases. These changes help bolster the global production of maize and prevent it from becoming unsustainable. Farmers and scientists can easily respond to the proliferation of pathogens growing on millet crops by leveraging the knowledge of the effect of the fungi on the plant. Disease-resistant corn strains, continuous crop rotation and disease control measures such as fungicides are recommended to farmers who can ensure that the effects of fungal diseases are reduced and therefore that maize yield will improve. Through research into the links between fungal diseases and decreases in maize yield, we can achieve food safety, economic viability and sustainable management of our ecosystem. In this way, key actors can learn and solve the vulnerabilities caused by fungal infections that threaten the output of maize and hence secure its future through the availability of this crop at all times [1].
Maize physiology and anatomy
Key physiological processes of maize growth and development: Maize, also known as Zea mays, is one of the most important cereal plants worldwide, supporting the population of food, including animals, distributed throughout the entire industry sector. As it unfolds, its growth and development are regulated by an impressive collection of microscopic physiological processes that enumerate its odyssey from a mere seedling all the way to harvest. In the course of this review, we provide a stage by-stage explanation of the mechanism underlying the growth and development of maize, which involves several fascinating processes.
Physiological processes driving maize growth and development
Germination and seedling establishment: Germination marks a stage where a maize seed wakes up from dormancy and is actively brought to life. Factors such as the temperature of the soil, moisture and oxygen availability greatly influence the percentage of seeds that germinate and the rate of growth during infancy. When the radicle finally moves and starts rooting and the plumule also pushes out, leading to shoot growth, the seedling does not stop and this process continues, leading to its establishment.
Photosynthesis and carbon assimilation: Photosynthesis, which is the heart of plant functions, involves the uptake of carbon dioxide and water from sunlight to produce glucose and oxygen, which are used to fuel maize growth. The interwoven combination of chlorophyll, stomata and the Calvin cycle has the ability to initiate the process by which maize plants manufacture carbohydrates and initiate the development of both vegetative and reproductive parts. Understanding the complexities of photosynthetic efficiency can be used predominantly for achieving optimum maize yields under different atmospheric conditions.
Water uptake and transpiration: Maize, a plant such as the rest, depends on a periodic uptake of water to create turgor pressure and help in nutrient transport and maintain its metabolic operation. The architecture of roots, water flow through them and regulation of stomata make it possible to adapt to major stresses, namely, drought and waterlogging, in maize. Deconstructing the processes governing water dynamics in maize plants provides access to response-gathering tricks for further modification of maize properties, including improved drought tolerance and improved water use efficiency.
Nutrient acquisition and assimilation: The consumption and metabolism of vital nutrients are fundamental processes required for maize to accomplish its growth and development goals. Root structure and symbiotic actions with soil microbes are two fundamental processes in nutrient absorption, whereas transportation systems across internals involve nutrient transfer from one part of a plant to another. The healthy growth of maize crops while maintaining their nutritional content is the major responsibility, indicating the role of effective fertilization programs in agronomy.
Hormonal regulation of growth and development: Maize hormones regulate diverse physical processes as mediators, controlling the division, growth, differentiation and death of cells. The roles of auxins, cytokinins, gibberellins, abscisic acid and ethylene in the modulation of environmental cues in plants could facilitate the physiological processes necessary for plant survival under stressful environmental conditions. Dissecting hormonal signal transduction routes reveals new ways to control a maize genetic makeup to gain traits that satisfy the farming needs of a contingent time.
Woven is a tiny thread of physiological processes; hence, maize growth and development are the major masterpieces of plant biology. There are as many as a billion such interactions, ranging from germination all the way to senescence, in which every step in the maize growing season is modulated by an abundance of environmental signals, hormone secretions and metabolic pathways. Molecular and biochemical studies of maize anatomy and structure have allowed researchers to develop new technologies aimed at increasing maize yield, resistance to climate-related threats and nutritional levels, which are vital to food security worldwide facing various changes [2].
Anatomical features of maize relevant to fungal pathogen infection
Corn (Zea mays is a lifelong crop worldwide and it provides substantial nourishment for humans and animals. In addition, it has applications in many industries and is an economic stabilizer. On the other hand, the productivity of transgenic wheat is limited by fungal pathogens, which, in turn, abuse anatomically weak nodes and vulnerabilities of this plant. In this review, we perform an extensive analysis of maize botanical aspects that reflect the pathways and pathogens involved in the susceptibility of various plant organs to pathogens.
Root system: The maize root system represents the primary soil feeding source and its main avenue within maize soil soil interactions while providing vital functions such as nutrient uptake, water absorption and anchorage. However, the fine carbuncular ground of the root system is also crossed by pathogenic fungi that penetrate and colonize corn. Root hairs, lateral roots and root exudates serve as microhabitats for fungal species colonization, while root hairs are important, as in the case of fungal pathogens such as Fusarium and Pythium, which act as entry points. Understanding the structure of roots and their interactions with fungal nemeses is pivotal for designing strategies that can accelerate root health and increase resilience.
Stem: The stem of the corn acts as a wire tree and other plants need to spread water, nutrients and assimilates further down the plants. However, the attack modes of pathogens such as Colletotrichum graminicola and Fusarium verticillioides are different from each other, as they cause diseases such as stalk lodging by taking advantage of the weakness in their stem anatomy rather than lowering them. Vascular bundles, pith tissues and nodes become infection sites that allow the fungus to enter. As they break into tissues, they cause vascular wilting, tissue necrosis and plant collapse. Researching stem characteristics and fungal pathogens is relevant for breeding resistant varieties of maize as well as promoting sustainable agriculture through cultural practices.
Leaves: The maize leaf is one of the main organs that conducts photosynthesis, transpiration and metabolic activities, which are crucial for plant development. At the same time, these genetic features of the varieties may also be very important for rendering plants susceptible to saprophytic fungi such as Exserohilum turcicum and Bipolaris maydis. Leaf stomata, trichomes and epidermal cell layers supply entry points and grooves where spore germination and hyphal spread occur. Understanding the leaf anatomy of maize in conjunction with the functional partnership dynamics of fungal pathogens is fundamental for developing an integrated disease management technique to prevent the destruction of maize leaves.
Ear and kernels: The ear and kernels correspond to the reproductive organs of maize and are key elements for pollination and fruiting processes. Nevertheless, such microbial groups of Aspergillus flavus and Fusarium graminearum are resistant to fungal pathogens that result in diseases such as ear rot and kernel mold. Certain botanical determinants, such as silk channels, husk coverage and cracks on kernels, serve as environments that encourage the formation of mycotoxins and fungal growth. Disclosing the anatomy of ears and kernels while considering fungal resistance properties is indispensable for conserving grain quality and security in maize production systems [3].
The morphology of maize plants is highly involved in facilitating hostile interactions of this fungal pathogen that can be encountered by different parts of the plant. With respect to the anatomy of roots and kernels, the fungus faces its own developmental challenges and can cause disease. By revealing the detailed mechanism of the anatomy of maize in interactions with fungal pathogens, investigators can develop focused tactics for disease control, line improvement and agricultural techniques so that adequate production and food safety can be achieved.
Interactions between maize physiology and anatomy in defense against fungal pathogens: Maize (Zea mays) is the major food crop worldwide and its production is often susceptible to pathogens. The physiology and structure of maize are similar to those of soldiers, which are the front line of defense against fungal infections. In this review, we will discuss the distinct correlation between the physiology and anatomy of maize plants, which allows them to develop the ability to fight fungal pathogens while at the same time illuminating underlying aspects of the mechanisms and way forward for crop improvement.
Physiological mechanisms
Hormonal signalling pathways: To address emerging fungal pathogens, hormones direct the production of a variety of defenses in maize plants through intra-actions. JA, SA, and ET are the key pathways in plants because they may activate defense related genes, which aid in the production of antimicrobial substances and the regulation of the cell suicide process. Through interactions between hormone-related pathways, maize can correct its antifungal reactions specific to targeted fungal pathogens.
Reactive Oxygen Species (ROS) production: From their genetic weaponry, maize plants quickly launch an oxygen burst, which includes superoxide radicals, hydrogen peroxide and different fungal species, as part of effective defense against fungal invasion. ROS harness themselves as signalling molecules, resulting in subsequent defense responses, including cell wall reinforcement, programmed cell mortality and activation of defense-related genes. The ROS-modulating property of maize is demonstrated by showing how maize either becomes more resistant or susceptible to fungal pathogens. These findings further show the importance of ROS homeostasis in plant immunity.
Anatomical adaptations
Cuticle and epidermal structures: The maize cuticle along its surface layer is made up of waxes and cutin that function as a physical barrier against fungi, thus restraining pathogenic entry into the inner tissue layers. Furthermore, dermal structures, such as hair and cells called papillae, form a layer that prevents fungal attachment, thereby reducing the chances of infection. Disease-resistant maize types with thick cuticles that form a protective layer and dense epidermal issues appear to be markers of the significance of anatomical adaptations in disease defense.
Cell wall composition and integrity: The maize cell wall acts as a primary line of defense against fungal pathogens and comprises complex polysaccharides, proteins and lignin. Alterations in cell wall composition, such as increased lignification or deposition of callose and cellulose, reinforce cell wall integrity and impede fungal penetration. Maize mutants deficient in cell wall synthesis enzymes often exhibit heightened susceptibility to fungal pathogens, emphasizing the critical role of cell wall architecture in disease resistance.
Integration of physiology and anatomy
Defense-related metabolite production: Maize physiology and anatomy converge in the biosynthesis of defense-related metabolites, including phytoalexins, phenolics and antimicrobial peptides. These metabolites are synthesized in response to fungal elicitors, pathogen-derived molecules that trigger defense signalling pathways. The spatial distribution of metabolite synthesis within different maize tissues influences the plant's ability to combat fungal pathogens, highlighting the interconnectedness of physiology and anatomy in defense responses.
Vascular transport and Systemic Acquired Resistance (SAR): The vascular system of maize serves as a conduit for the transport of defense signals and metabolites throughout the plant, facilitating Systemic Acquired Resistance (SAR) against fungal pathogens. Upon localized pathogen attack, signalling molecules such as SA and jasmonates are transported systemically, priming distal tissues for enhanced defense responses. The integration of vascular transport mechanisms with physiological and anatomical defenses confers broad spectrum resistance against fungal pathogens in maize.
The intricate interplay between maize physiology and anatomy lies at the heart of its defense against fungal pathogens. From hormonal signalling pathways to anatomical adaptations, maize plants deploy a diverse array of strategies to fight fungal infections and maintain crop health. By unravelling the complexities of this interplay, researchers can uncover novel targets for crop improvement, paving the way for sustainable maize production in the face of evolving fungal threats.
Maize genetics and resistance mechanism
Major Quantitative Trait Loci (QTLs) associated with fungal pathogen resistance: Maize, a key cereal crop, experiences several constraints from fungal diseases that cause significant losses in yield and grain quality. Knowledge of the genetics that confers resistance is a key factor in the production of superior maize varieties. Quantitative Trait Loci (QTLs) are an integral part of the genetic breeding process.
QTLs are chromosomal regions that contain genes that constitute genes with a particular trait, e.g., resistance to fungal diseases. Fungal diseases such as northern leaf blight (caused by Exserohilum turcicum) or gray leaf sport (caused by Cercospora zeae maydis) can damage maize crops. Many QTLs related to resistance to these diseases and other fungal diseases have been identified.
The QTLs frequently cover more genes, each of which has a low impact on disease resistance. The expression of these genes is consistent with the general level of resistance, which is typical for a particular corn variety. The linkage mapping of QTLs implies the genome transfer of resistance traits and genetic markers across generations [3].
The how QTL analysis aids in breeding for resistance
Targeting breeding efforts: By identifying the QTL regions responsible for the resulting genotypes, breeders can concentrate on the selection of plants with the most appropriate QTL alleles within breeding programs.
Marker-Assisted Selection (MAS): DNA markers linked closely to resistance QTLs that can be used for early selection of resistant plants during the growing season can be applied, overcoming the resource-and time-consuming implementation of disease phenotyping.
Gene discovery: Modern techniques such as positional cloning may be used to specify the genes in the resistance-related QTLs. Such knowledge is also instrumental in increasing resistance through gene modification or gene pyramiding of multiple genes from different pathogens for greater protection.
Challenges and future directions: Despite these difficulties, QTL analysis still has an important opportunity. The complex involvement of resistance genes, ranging from epistatic to environmental interplay with QTLs, often makes it difficult for the gene discovery process to follow through. Furthermore, QTL effects on the pathogen in question are variable and may depend on the concrete variety as well as the actual environmental conditions.
Future research directions involve the following:
•In addition to the more precise mapping of QTLs and identification of related technologies will be employed. genes, modern sequencing
•QTL mapping across diverse panels can be combined to improve our understanding of how disease resistance genes function.
•Classic approaches such as QTL mapping are combined with the latest approaches to produce genome editing techniques to promote broad and stable resistance in maize.
By undertaking more research to genetically fingerprint complex fungal diseases in maize, researchers can equip breeders with breeding material to create varieties that are highly resistant and robust to these diseases as the world population continues to grow and there is a pressing need to grow more food.
Candidate genes and their roles in resistance mechanisms: In addition to QTLs, researchers are narrowing candidate genes in regions that play a critical role in maize defense against fungal pathogens. These genes are responsible for several proteins involved in the plant immune response, including Pathogenesis Related (PR proteins, enzymes that may eliminate toxic materials and signal transduction components. Recognizing the position of these genes and their implications for fungal virulence factors is important for developing confirmed methods to strengthen maize resistance. Candidate genes are genes that are likely involved in resistance mechanisms [4].
ZmNPR1: The immunity gene that provides systemic acquired resistance as a response pathway.
ZmbZIP: A transcription factor that turns on defense-related genes.
ZmPR1: This gene is translated into a pathogenesis-related protein with antifungal function.
Defense signalling pathways: Defense signalling pathways are crucial for activating resistance mechanisms in maize. The Salicylic Acid (SA) pathway and the Jasmonate (JA) pathway are two key defense signalling pathways. The SA pathway regulates systemic acquired resistance, while the JA pathway regulates induced systemic resistance.
Maize utilizes a complex network of signalling pathways to recognize and respond to fungal pathogens. These pathways involve a cascade of events triggered by the perception of Pathogen-Associated Molecular Patterns (PAMPs) by plant cell surface receptors. This perception leads to the activation of Mitogen-Activated Protein Kinase (MAPK) cascades, which in turn induces the expression of defense genes and the production of various defense compounds.
Physical barriers and antifungal compounds: Usually, several measures are taken to establish a defense against fungal pathogens that is physical and antifungal.
The plant cell wall acts as the first guardian, where cellulose and lignin are the two main components of mechanical opposition to fungal invasion. In addition, grain has highly developed biosynthetic capacities, which allows it to produce a plethora of fungicidal compounds, including phenolic compounds, pathogenesis-related proteins and secondary metabolites. These substances can slow fungal growth, destroy fungal cell membranes or decrease the number of generated radicals.
For example, physical barriers such as cuticles and cell walls prohibit fungal entry. Flavonoids and terpenoids function as antifungal agents and they arrest the growth of fungi. Genes such as ZmPAL and ZmTPS, which are associated with the biosynthesis of these compounds, are responsible for the efficacy of the resistance mechanism.
Gene expression modulation: Orchestrating defense: Gene expression modulation in maize-fungal pathogen interactions: In this situation, maize vividly indicates that the gene should be dynamically managed in the context of fungal infection. The entire execution of such a process occurs on a delicate level (because) it is a complex mix of several signalling molecules and transcription factors that act in synchrony. The recognition of PAMPs by maize cell surface receptors is the initial step of a mechanism that leads to the activation of a signal cascade. In most cases, this process is accompanied by processes known as MAP kinase cascades. MAPKs in these pathways are activated and then function as passers to the phosphorylation of downstream transcription factors. In this way, several phosphorylated transcription factors overexpress different defense genes by binding to promoter DNA sequences and transcription to mRNA, with further translation to functional proteins [5].
The chosen genes and signalling cascades are influenced by the nature of the fungal pathogen and the plant's defense scheme. In one example, the recognition of the particular molecular pattern responsible for fungal recognition would trigger the expression of genes involved in the production of PR proteins, enzymes that would help in the detoxification of fungal microtoxins or molecules responsible for cell wall strengthening. The accuracy with which this molecular machinery customizes gene expression allows maize plants to have a specialized defense against particular fungal invaders.
Several key components are essential for this gene expression modulation
Signal transduction molecules: MAPKs are different messengers that transduce signals from PAMP recognition to the nucleus, where they activate gene expression and initiate a defense reaction.
Transcription factors: These proteins specifically attach to certain DNA sequences in gene promoters, which could lead to an increase or decrease in the rate of gene transcription.
Defense genes: These genes produce proteins as well as enzymes that are biochemically important for different mechanisms of plant immunity, including PR proteins, antifungal compounds and oxidative stress enzymes.
It is of great importance to unravel the complexities of gene expression turnover in fungal pathogens so that we can develop novel control strategies for diseases. Researchers have done this by singling vital regulator genes as well as their protein proteins and then, they proceeded to explore the probability of manipulating pathways that improve maize resistance.
This might involve: Engineering plants with constitutive activation of defense pathways. This can provide broad-spectrum resistance as a multitarget against different fungal pathogens.
Developing fungicides that target specific steps in the fungal pathogenesis process: Via disrupting fungal PAMPs or attenuating fungal downstream signalling pathways, such molecules could abate the ability of pathogens to sway plant gene expression.
The technology behind developing resistance mechanisms is gene regulation. ZmWRKY and ZmMYB are transcription factors that are involved in regulating defense gene expression. On the other hand, DNA methylation and histone modifications, which are epigenetic mechanisms, have attracted much attention as factors that modulate gene expression.
In conclusion, genetic expression modification is the most crucial factor in maize defense against viral fungal pathogens. The whole process by which chemical and transcription factors work together to ensure the resistance of maize varieties is highly complex. Breeding this process opens the door for new ways of breeding maize that will enable people to easily face fungal threats that are evolving at an alarming rate.
Breeding strategies for developing resistant maize varieties
Breeding strategies for durable resistance in maize: Battle with fungus invasion: Maize pathogenic fungi represent a significant threat to maize production, causing yield losses and reducing grain quality. To address this obstacle, plant geneticists are resorting to a wide gamut of techniques that will give rise to maize varieties with improved resistance [6]. This section is dedicated to explaining some of the most important breeding methods for successful resistance development in corn to fungal parasites.
Conventional breeding methods
Selection for natural resistance: This customary approach targets competitive maize germplasm selection for those cultivars exhibiting resistance against particular fungal diseases. These particular resistant lines were later used as parathions in breeding programs to pass resistance traits into new varieties. This way of doing things is quite simple, but its execution may be time-consuming and rather constrained by just how much naturally efficient germplasm is in supply.
Hybridization: In trying to make the most out of the process of heterosis, breeders combine elements of genetically distinct parental lines to create F1 hybrids that have high disease resistance, high yield and other top-notch features. This method is widely employed in commercial corn production, but this process necessitates breeding to maintain heterosis (universal hybrid vigor) for subsequent generations.
Recurrent selection: This approach consists of generating populations that sequentially undergo cycles of selection for a trait of interest (disease resilience) and intercrossing with the target individuals. This method involves harnessing this allelic concentration to lead to overall population resistance, which takes multiple generations to evolve. Repeated selection is the most common absorbance type that can generate resistance to complex diseases with polygenic inheritance.
Marker-Assisted Selection (MAS): In recent years, the use of molecular markers has completely altered classical plant breeding to a stage in the indirect selection of genes linked with competitive traits. Within the framework of MAS (Marker Assisted Selection), DNA markers that cosegregated with Quantitative Trait Loci (QTLs), which are inferred by linkage mapping, were used. Breeders find it more convenient to use markers to identify and select breeding populations that harbor desirable disease resistance genes even before the season starts and without the need to waste time studying physical disease symptoms. Such rapid-cycle selection is highly effective for fast tests and resistance traits.
Combining conventional and modern approaches: Modern breeding correlates the conventional approach with sophisticated methods such as MAS. In this way, breeders are given the leeway to use the suitable trait of each method. Another example can be the joint utilization of conventional breeding and MAS, where valuable lines are located upon their resistance potential and tagged QTLs related to resistance are then used for targeted selection. This is an effect of combining these methods, as they can result in the production of competent maize seeds that are resistant to both insects and long-lasting diseases [7].
Emerging technologies and future directions
Genome editing: Methods such as CRISPR-Cas9 make it possible to achieve specific modifications that are related to the resistance of genes. On the one hand, breeders may choose to selectively edit certain genes within a maize variety to devise new resistance mechanisms or improve the existing ones; on the other hand, they have the potential to fix this issue.
Speed breeding: This forward-thinking method harvests accelerated life cycles, in conjunction with the use of an environmentally controlled system, to create multiple generations of plants much quicker. This is a key factor, as it is faster to develop new varieties with improved resistance traits by contrasting traditional farming systems.
Breeding for multipathogen resistance: In fact, the wide spectrum of complex fungal pathogens that are predisposed to one pathogen may not persist long. In the future, the use of breeding filtrates could be aimed at developing varieties with broad-spectrum resistance or ultimately pyramiding resistance genes for several diseases caused by fungi.
Challenges and considerations: The development of durable resistance in maize requires careful consideration of several factors. The dynamic nature of fungal pathogens: Fungi pathogens are subject to rapid adaptations that make them immune to known resistance genes. Breeders must improve the breeding of varieties with various types of resistance as the race between resistance mechanisms develops in the evolutionary arms race.
Environmental influence: The efficiency of resistance traits can change easily if certain environmental factors, such as temperature, humidity and nutrient availability, are involved. The compatibility of resistance genes is highly important for breeding programs that aim to maintain resistance to diverse kit growing conditions.
Integration with other agronomic traits: Resistance, though critical, should not affect other important agronomic traits, including yield, maturity and grain quality. Breeders are trying to produce cultivars with a mix of advantages that provide a balanced combination.
Unveiling the maize pathogen reaction–a multiomics approach
Genomics: Deciphering the language of resistance.
Identifying resistance genes and mutations: Plants are often considered the lower form of life and are neglected in research and support. On a daily basis, plants face major challenges such as fungi, bacteria, viruses and pests. To protect themselves from the attackers that are intended to harm them, plants have evolved complex and diversified defense systems but with the use of resistance genes. In this review, we explore the complex universe of plant resistance genes and mutations, revealing the methods and tools used to identify and exploit the expression of these genes related to immunity.
Nucleotide-Binding Site-Leucine-Rich Repeat (NBS-LRR) proteins: R-genes such as NBS-LRR-type genes are arguably the most quantitative and most scattered class of plant resistance genes that provide protection against diverse kinds of pathogens. The majority of these proteins usually have two distinct domains, namely, a Nucleotide-Binding (NB) domain and a Leucine-Rich Repeat (LRR) domain, which are responsible for pathogen detection and activation of defense responses. The identification of both NBS and LRR genes mostly occurs through GWASs and transcriptomics, which were subsequently validated through the use of genetic transformation or gene editing approaches [8].
Receptor-Like Kinases (RLKs) and Receptor-Like Proteins (RLPs): Moreover, RLKs and RLPs carry members of this group of resistance genes that are involved in pathogen recognition and signal transmission. These pattern recognition receptors are typically of the membrane-bound type and they have the ability to activate pathogen-associated molecular patterns or pathogen effector molecules that stimulate an immune response. Identification of RLKs and their Ligand Proteins (RLPs) requires a combination of bioinformatics, transcriptomics and protein protein interaction analysis, which should later be complemented with knockout and methodologies to reveal their precise functions.
Resistance (R) gene clusters: In several plants, as a defense mechanism, genes involved in resistance are located in certain zones of the genome, called R gene clusters, which quickly evolve in response to pathogenic attacks. The observed patterns of differentiation can be ascribed to selective sweeps and population bottlenecks; these local adaptations often yield significant allelic variability. Identifying and characterizing R gene clusters include comparative genomics, haplotype analyses and population genetics; thus, etiologic patterns emerge concomitantly with plant-disease interaction development.
Harnessing mutations for enhanced resistance
Induced mutagenesis: Induced mutagenesis methodologies such as chemical mutagenesis and radiation mutagenesis have been helpful in advancing the creation of genetic diversity through controlled and expressed mutagenesis for improving crop traits. Populations generated via mutagenesis approaches are screened for favourable characteristics, such as stronger fight against disease-causing organisms. NGS technologies are instrumental for the rapid identification of mutations that are responsible for resistance traits and greatly reduce the risk of unintentionally creating undesirable traits by using markers in the breeding process.
Genome editing: Genome editing technologies, such as CRISPR-Cas9, represent the advancement of this science by permitting one to introduce or modify mutations in plant genomes with more exactness and targeting. Through the addition, deletion or alteration of single nucleotides, genome editing can change alleles into novel alleles or alleles linked to pathogen resistance genes; therefore, if individuals use gene therapy, they will become highly resistant to pathogens. The specificity and rapid rate at which CRISPR-based techniques confer resistance to mutations hinder the discovery and use of these disease-resistant cultivars in crop breeding processes.
Functional validation: There is a limitation in the linking of functional genes and mutations in genomics technology despite advances being made. The process of crop improvement was stopped. Intact phenotyping platforms and transgenic techniques are required to identify the molecular components that lead to resistance traits and to assess the efficiency of such traits in field trials [9].
Deployment of multigenic resistance: Many plant pathogens exhibit genetic variation that can be very high and they can develop resistance to genetic forms of resistant plants very quickly. Consequently, in addition to having these single genes, stacking multiple resistance genes or Quantitative Resistance Loci (QTLs) should be applied to sustain disease control.
Genome-Wide Association Studies (GWAS): Genome-Wide Association Studies (GWASs represent an advanced tool for geneticists, offering a way to explore the genetic mechanism underlying complex traits and diseases across a wide array of species. Genome-Wide Association Studies (GWASs provide researchers with the ability to investigate millions of genetic variants spread across the whole genome. As a result, it becomes easy to identify genetic loci that are associated with the phenotypic variation present in populations. This review discusses the theories, strategies, use cases and challenges of GWASs, introducing their role in the discovery of gene and phenotype relationships.
Principles of GWAS: GWAS implements the principles of LD and allelic association to identify the source that influences phenotypic traits. HLA is a generic name for a set of alleles that are not distributed evenly within a population and that undergo recombination. GWAS can employ long-range LD patterns to determine marker trait associations, which designate specific DNA variants (for example, Single Nucleotide Polymorphisms (SNPs that are statistically correlated with traits of interest.
Study design and population selection: Regardless of the chosen approach, (the most common is GWAS will often have to assemble a large cohort of populations with representative genotypic/phenotypic variation of interest. Various meticulous approaches have been adopted in relation to population structure, relatedness and environmental variables to reduce the effects of confusing factors on the associations of genetics. The structure of a population can be analysed via methods such as Principal Component Analysis (PCA and relatedness can be determined through kinship analysis.
Genotyping and quality control: By way of microarrays and Next-Generation Sequencing (NGS, we are able to perform genotyping of individuals via genetic markers across the genome at a higher throughput level. The quality control process, which aims at filtering out low-quality markers and samples and should genotypic data be used for downstream analysis, is implemented and the results are highly reliable. Scientists can perhaps incorporate other approaches, such as imputation, that can be used to fill in where there are no markers using DP patterns.
Statistical analysis: Linear Mixed Models (LMMs or logistic Regression Models (LRMs are commonly used for identifying associations between markers and traits. In addition, Generalized Linear Models (GLMs are also very common.
Applications of GWAS: GWASs assist in pinpointing genomic loci according to traits such as agronomic and pharmacogenomic response profiles. Candidate gene markers for GWASs offer a greater rank of significance than others because they carry information about how the trait is modified. The next phase is functional validation. Through GWAS, thousands of genetic variants that are associated with human height, BMI and common disease development have been identified [10].
With respect to crops, GWAS findings, in turn, help breeders with MAB to identify and introgress anb2te better alleles in other crop breeding programs. In MAS, the pools include the trait loci identified in the GWAS that are linked to markers.
Marker-assisted breeding expedites the progress of these positive traits. GWAS, for instance, enabled the discovery of gene variants with a profound effect on drug uptake, disease susceptibility and personalized treatment approaches in the field of medicine.
Nevertheless, despite the advantages of GWASs, there are some conflicting areas, such as the need for large sample sizes to detect small-effect variants, accounting for multiple confounding factors and interpreting noncoding variants. Undoubtedly, the development of new GWAS methods, such as multiplex genomic exhaustiveness and functional genomic and single-cell genome sequencing, has the capacity to increase the precision of genetic connections.
Transcriptomics: Unmasking the gene orchestra during infection
Understanding gene expression changes in response to stress: In the constantly evolving realm of living organisms, creatures are exposed to ecological challenges that destabilize their metabolic processes and threaten their existence. This complex molecular system, which is analogous to orchestra, functions as a medium of adaptive response, acting to modify gene expression patterns to counteract the harmful effects of stress. We focused on the fascinating gene expression changes induced by stress and investigated how the underlying molecular mechanisms underlying adaptive responses may differ across species.
Molecular basis of stress response
Perception and signal transduction: This process first begins when facilitating intracellular function via environmental sensing or through specialized sensor-receptor pairings, which then initiates signal transduction cascades. Therefore, signalling pathways, including Missing-Activated Protein Kinases (MAPKs) and calcium-related pathways, send stress information from the cell membrane to the nucleus, which affects gene expression. While Transcription Factors (TFs) are the core components of stress responses and are the top candidates for binding regulatory elements in target genes, they are the key players that convert stress signals into changes in gene expression patterns, which will be discussed in the following section.
Transcriptional regulation: Stress-Inducible Transcription Factors (TIFs), such as Heat Shock Factors (HSF), Dehydration Responsive Element-Binding Proteins (DREB) or Abscisic Acid Responsive Element-Binding Factors (AFB), undergo transcriptional reprogramming in response to stress. These TFs modulate the expression of stress-responsive genes involved in various molecular pathways that help plants endure stressful conditions and detoxify and repair damaged tissues. In addition, epigenetic alterations, such as DNA methylation and acetylation of histones, restrict gene expression by influencing chromatin accessibility in the face of stress.
Functional genomics approaches
Transcriptomics: Gene expression profiles are generated using transcriptomic profiling, which is a technique involving RNA sequencing (RNA-seq) and microarrays and a wide genomic view of changes in gene expression in response to stress is obtained to provide insight into gene implications under these conditions. Transcriptome data comparison involves identifying transcriptional networks that are dynamic and revealing stress induced gene pathways. Interlaced experimentation (e.g., time course represents the dynamic nature of changes in gene expression as an early response, late responsive genes are revealed and regulatory mechanisms are revealed.
Proteomics and metabolomics: The integration of transcriptomic data with other datasets, such as proteomic and metabolomic data, provides a complete picture of cellular processes responding to stress. Proteomic techniques, such as mass spectrometric proteomics, can detect variations in protein quantification, posttranslational modifications, and protein protein interactions in response to stress. Metabolomics has led to the discovery of metabolite content changes and abnormal metabolic pathways that contribute to the stress response. These findings may help to elucidate how cells adjust their metabolism during stressful conditions.
Functional insights and applications
Stress tolerance mechanisms: An array of different stress tolerance mechanisms is built on alterations in gene expression. These factors include osmotic adjustment, antioxidant defense and protein quality control. The stress-responsive genes of proteins that act as molecular chaperones and provide osmoprotection and detoxification address the breakdown of cell homeostasis in the case of stress. Addressing the molecular principles of stress tolerance enables the genetic improvement of plants that, together with the amelioration of the environment, contributes to sustainable agriculture.
Biomarker discovery: An organism's preservation, capabilities and susceptibility to stress are all determined by stress-responsive genes and molecular signatures. Biomarker-based research uses transcriptomics and proteomics datasets to search for biomarkers that can be correlated with stress conditions or individual physiological states. These biomarkers may be used in environmental monitoring, agriculture and human health, thus allowing for early detection and repelling of stress-related hazards through interventions.
Many studies have been conducted, but there are still challenges associated with decoding the intricacies of gene expression changes in response to stress. Context-specific response deciphering, deciphering the regulative network of gene sorting, and determining gene function when stressed are the core of ongoing research. Multiomics data integration, predictive modelling techniques and advanced imaging approaches are very promising because of increased knowledge about the biological underpinnings and resilience mechanisms of stress.
Identifying key regulatory pathways: Infection represents a core development of relationships among pathogens and the host organism to set off intricate biochemical processes to fight invasion and maintain homeostasis. A key factor of molecular symphony is this intricate network of regulatory pathways, which direct the transcription of genes related to immune reactions, pathogen identification and tissue regeneration. In this review, we move from the description of the various pathways that usually recognize regulatory genes to the music of the gene orchestra during infection, thus unveiling the dance of molecules that takes place inside infected hosts [11].
The infection process involves PRR sensing of PAMPs that are secreted by pathogens such as TLRs, NLRs and RLRs. PRR activation transduces intracellular signals, precipitating the expression of key downstream molecules, i.e., the NF-κB, MAPK and IRF pathways, which control the transcription of immune response genes. These routes are essentially the central junctions where extracellular signals and intracellular signals are united and immunological responses are coordinated to defend against infection.
Cytokine and chemokine signalling: Cytokines and chemokines function as progression molecules by inducing immune cell mobilization, activation and effector performance during infection. Upon binding to receptors such as the JAK STAT and MAPK pathways, key cytokine subsets such as Interleukins (ILs), Interferons (IFNs) and Tumor Necrosis Factors (TNFs) activate various signalling pathways such as the JAK-STAT and MAPK pathways. Dysfunction of cytokine regulation may be associated with the risk of inflammatory disorders, autoimmunity or increased susceptibility to infections.
Identifying regulatory pathways
Transcriptomics and systems biology: Transcriptomic profiling by employing high-throughput sequencing technologies, including RNA-seq, leads to a gene expression puzzle of infection. The use of transcriptomics techniques such as Gene Set Enrichment Analysis (GSEA) and network analysis will help in obtaining deeper insights into how regulatory pathways and Gene Regulatory Networks (GRNs) function in host interactions.
Genetics: Genomic functional techniques, such as CRISPR mediated gene editing and RNAi, allow investigators to control the modification of gene expression levels to uncover the functional role of regulatory pathways in the course of infection. Genome-wide profiling screens, including CRISPR knockout screens and RNAi screens, have identified genes and pathways that play a role in host defense or pathogenicity. In conjunction with transcriptome data, functional genomics methodologies reveal more information about the regulative logics governing the processes and responses within infection.
Dynamic regulation of immune response genes: Regulation involves the expression of immune response genes, such as cytokines, chemokines, antimicrobial peptides and immune receptors, in a simultaneous and coordinated manner. The transcription factors NF-κB, IRFs and AP-1, which are triggered by pathogen-sensing gene expression, are activated or repressed. Coordinated reactions to immune response genes, together with proper functioning of these genes, result in clearance of infection and elimination of inflammation [12].
Crosstalk between regulatory pathways: Regulatory pathways include over and back-interrelations and feed-forward loops, which determine the intensity and duration of immune responses once infections develop. Cross-responsibility among pathways, such as the NF-κB and STAT pathways, is necessary for the immune response to be set to a point where the pathogen is cleared manually while tissue preservation and immune tolerance are achieved. Distortion of the communication system composed of these mechanisms may result in disordered functioning of the immune system or chronic inflammation, showing that there is great significance of system integration in infection.
Proteomics: Unveiling the protein players in defense
Analysis of protein expression patterns and functions: Proteins represent the very action force, the staff behind the scenes responsible for almost anything done by the organism. To elucidate cellular machinations, pathologies and treatment options, the foundations of protein expression and function are key ancillary to discovering the intricacies of cellular physiology and disease. This article details the techniques and uses of protein expression patterns and functional analysis, which will supplement the readers’ knowledge and appreciation of the seemingly intricate processes that underlie the dynamics of cells.
Analysis of protein expression patterns
Proteomics technologies: The term “proteomics” covers a group of techniques intended for the investigation of the entire set of proteins belonging to a certain biological system. Proteomics utilizing mass spectrometry methods, including shotgun proteomics and targeted proteomics, will enable large-scale protein identification and quantification of proteins in complex samples. LC-MS/MS (Liquid Chromatography coupled with tandem Mass Spectrometry), on the other hand, deepens the monitoring of protein profiles and Posttranslational Modifications (PTMs); hence, insights into the network of signalling and regulatory mechanisms are available.
Protein profiling techniques: Protein quantification methods, including western blotting, IHC and ELISA, are extremely useful techniques because they allow scientists to investigate alterations in a specific set of proteins in biological samples. Western blotting makes it possible to measure the quantity and quality of proteins by antibody detection. At the same time, IHC can be used to determine the site of proteins that are also linked to uniquely specific antibodies. The ELISA technique detects the presence of a protein in biological fluids; thus, it has the highest level of sensitivity and specificity, providing researchers with useful data for biosignatures discovery and diagnostics.
Functional characterization of proteins
Functional genomics approaches: The evolution of functional genomics based on gene knockout, RNA interference and the CRISPR Cas system has been carried out to study protein functions in both cells and entire living organisms. Knockout studies include the deletion or disruption of selected genes in model animals, during which their appearance is investigated according to the loss-of-function of the corresponding proteins. RNA interference (RNAi) allows gene expression to be knocked down transiently or stably by RNA-mediated degradation, which provides the benefit of reversible evaluation of the implicated gene. With CRISPR, genome editing can be performed accurately by modifying genomic sequences through kick-out, knock-in or functional element regulation, allowing quicker and more precise modulation of protein function [13].
Functional assays and pathway analysis: Stable assays such as enzyme activity assays, proteinâ??protein interaction assays and cellular phenotypic assays provide information on protein biochemical characteristics and cellular mechanisms. Enzyme activity assays revealed the catalytic activity of proteins, revealing their function in metabolic pathways and signalling cascades. Protein-protein interaction binding tests and Coimmunoprecipitation (Co-IP) and Yeast Two-Hybrid (Y2H) assays revealed physical contacts between proteins, hence revealing protein complexes and signalling pathways. Cellular phenotypes are tested to determine the impact of protein perturbations on cellular processes with respect to proliferation, apoptosis and differentiation and to identify these motions as direct consequences of such protein dysregulation in disease states.
Applications in biomedical research and beyond
Biomarker discovery and disease diagnosis: Disease characteristic markers (biomarkers) can be identified via analysis of protein pattern expression and functions. Dysregulation of protein expression profiles in several diseases, such as cancer, neurodegenerative disorders and autoimmune diseases, is one of the well-known causes. A biomarker of a disease would enable early diagnosis, prognostic assessment and personalized treatments, which eventually improve a patient's outcome.
Identifying potential biomarkers and therapeutic targets: The presence of proteins plays a role in the readiness of organisms’ defense systems to fight several agents, such as disease-causing microbes, toxins and poor environmental conditions. The study of the entire proteome within a biological system, i.e., proteomics, is a significant approach toward identifying the unique role played by proteins in defense, not only because it can provide biomarkers but also because it can target substances that can be used for treatment. In this review, we describe how proteomics has deciphered the defensive system and attempted to identify hormonal markers and therapeutic targets for preventing illness and increasing resilience.
Proteomic approaches in defense research
Shotgun proteomics: Shotgun proteomics is the method of characterizing total protein expression in biological samples and is derived from high-throughput Mass Spectrometry (MS). Proteins are hydrolysed into peptides, which are later separated by liquid chromatography and further mass spectrometry analysis is conducted to spot and measure the amounts of individual protein constituents. Shotgun proteomics enables the global assessment of proteinomes and allows the observation of variations in Protein Modifications (PTMs).
Targeted proteomics: In targeted proteomics, the identification and quantification of particular proteins and their modifications at the isotype level are emphasized. Partnering with SRM or PRM assays is achieved. Targeted proteomics can be used because of its superior size, accuracy and ability to perform multiple simultaneous examinations of large amounts of data. The discovery-based proteomics approach can be used for identifying candidate biomarkers and identifying therapeutic targets [14].
Identifying biomarkers of defense
Disease biomarkers: These molecular indicators trace imbalances in an organism's state and determine whether disease is present, how far it has progressed or the response to treatment. Continuous progress in the realm of proteomics based biomarker discovery offers an opportunity to study differential protein expression patterns as well as posttranslational modifications to identify specific biomarkers associated with homeostatic mechanisms. The biomarkers of different diseases are early detection, prognosis and personalized treatment strategies, which make them indispensable tools for improving health outcomes and health care efficiency.
Host interactions: Proteomics contributes to understanding the molecular interactions between hosts and pathogens in the course of infection, among others, disease biomarkers, pathogen virulence factors and drug targets for antibacterial interventions. Proteomic profiles of infected and uninfected material can reveal protein expression, positioning and interactions related to disease course. The role of biomarkers related to the host immune response and pathogen virulence in the detection, therapy and prevention of infectious diseases has become clear.
Drug targets: Proteomics-driven target identification improves the chances of finding defense-related proteins and their related pathways that can be drugged. Proteomic analyses in which diseased and healthy tissues or cells are compared pinpoint the dysregulated proteins and/or signalling pathways that are related to disease development according to Guo and Li. Drug-able sites or places, such as enzymes, receptors and signalling molecules, are the primary forces used to alter the defense mechanisms and homeostasis of cells via medicinal responses.
Immunotherapeutic targets: Immunotherapies are designed to recruit the body's immune system to fight diseases such as cancer, autoimmune disorders and viruses. Proteomics expands the prospects of finding novel immunotherapeutic targets, e.g., tumor-specific antigens, regulators of immune checkpoints and immunomodulatory proteins, that function both in immune activities and in the tumor microenvironment. Since immunotherapeutic targets were discovered via proteomics, they are key for the development of novel immunotherapeutics that are more effective and have fewer adverse effects.
Metabolomics: Mapping the metabolic landscape of infection
Characterizing the metabolic changes in response to infection: Infection involves a metabolic transformation that begins in infected cells and then spreads through the tissues to support the immune system and the survival of the virus or bacteria. Host and pathogen metabolism studies via metabolomics, the comprehensive analysis of small molecules in biological systems, provide ideas about host-microbe interconnectivity dynamics, particularly in infectious disease development. This paper describes the techniques and uses of metabolic characterization in response to infection and tracks metabolic routes as the host battles the pathogen via metabolomics [15].
Untargeted metabolomics: Random metabolomics is conducted through the global profiling of metabolites in biological samples without any prior known data on their species. Using high throughput methods, such as Liquid Chromatography Mass Spectrometry (LCMS) and Gas Chromatography Mass Spectrometry (GCMS), it is possible to detect and quantify different molecules, including amino acids, lipids, carbohydrates and nucleotides. Through metabolomics, an indication of the overall metabolic disturbances linked to infection is revealed and this knowledge suggests the modes by which the host pathogen interactions are influenced and the mechanisms of disease are elaborated.
Targeted metabolomics: Targeted metabolomics is a form of analysis that emphasizes the quantitative determination of specific metabolites or individual metabolic pathways based on specific metabolite standards. For instance, the range of targeted metabolomics assays, which include highly sensitive and specific techniques such as Liquid Chromatography-Tandem Mass Spectrometry (LCMS/MS) and Nuclear Magnetic Resonance (NMR) spectroscopy, provides information about metabolite concentrations in biological samples. In addition, metabolomics enables accurate investigation of the most significant metabolites involved in host protection mechanisms, microbial metabolism and host-interactions during infection.
Mapping the metabolic landscape of infection
Host metabolic reprogramming: Infected hosts undergo more rigorous changes in metabolism to meet the energy needs of immune and repair systems. Metabolomics-based investigations have shown changes in glycolysis, the Tricarboxylic Acid (TCA) cycle, amino acid metabolism and lipid metabolism in affected host cells and tissues. Metabolic shifting feeds upon increased immune cell activation and the generation of reactive oxygen species, thereby impacting the number and function of antimicrobial elements. In this case, the metabolism of the body is altered.
Microbial metabolism: Human-unfriendly microbes parasitize host nutrients and metabolic pathways to increase their growth, virulence and survival in the host environment. Metabolomic analyses revealed bacterial metabolic adaptations during infection and revealed virulence factors, metabolic necessities and drug targets. Analysis of the metabolic makeup enables us to determine the mechanisms of nutrient uptake and metabolic routes, as well as to understand how microbes interact with their host, which in turn is of great significance for drug design and therapeutic procedures.
Applications in infectious disease research
Biomarker discovery: The application of metabolomics in proteomic biomarker discovery has led to the detection of metabolic spectra that are linked to infection, disease severity and treatment effectiveness. Metabolomic analysis revealed disease-specific metabolites in a range of fluid, tissue and ex vivo culture samples. Metabolic biomarkers can be applied for diagnosis and prognosis and cancer monitoring might also help clinicians detect early disease and stratify patients at risk, such as through individualized treatment plans [16].
Drug target identification: Metabolomics-inspired target discovery allows for the identification of enzymes, transporters and pathways of exceptional importance for microbe growth and survival. Metabolic profiling of pathogens can be performed if their different growth conditions or drug treatments are tested. This is how metabolic vulnerabilities and drug targets are revealed. The development of antimicrobial agents that disrupt metabolic pathways that function in pathogen-specific functions, such as cell wall biosynthesis, nucleotide metabolism and redox homeostasis, may result in the development of novel antibiotic therapies with broad-spectrum activity and reduced resistance.
Understanding how maize alters its metabolism for defense
Crop agriculture, one of the world's most important ongoing processes, is regularly under the threat of pests, pathogens and environmental stresses. For the sake of its survival, maize has developed complex defense mechanisms that consist of metabolic adjustments enabling these plants to react swiftly to a variety of threats. Understanding how maize metabolism changes for defense can be vital for improving crop quality and yield. In this review, the subtle and complex metabolic strategies of maize that enable it to mount defense responses in response to biotic and abiotic stresses are explored. This review provides insight into the underlying molecular mechanisms underlying maize resilience to biotic and abiotic stresses.
Phytohormone signalling: Maize damage begins at the level of stress perception, which initiates signalling cascades mediated by phytohormones such as Jasmonic Acid (JA, Salicylic Acid (SA and Abscisic Acid (ABA. These phytohormones, which are important signalling molecules, regulate defense responses. The Jasmonate (JA signalling pathway is the central pathway leading to the activation of genes involved in the production of defense related secondary metabolites such as terpenoids and phenylpropanoids, which confer resistance against herbivores and pathogens. The SA pathway influences the expression of PR genes and promotes Systemic Acquired Resistance (SAR, an immune mechanism against pathogens that is not specific. Among the mechanisms of stress tolerance, the ABA signalling pathway modulates stomatal closure, osmotic adjustment and antioxidant defense in maize plants under drought and salt stresses, increasing plant resilience.
Metabolite profiling: Metabolite profiling is a broad-spectrum study of the maize metabolome for revealing primary and secondary metabolic alterations occurring in response to environmental stress. The metabolic reprogramming of stressed maize plants results in the production of these defense compounds, notably flavonoids, alkaloids and phytoalexins, which serve as proteinaceous antimicrobial agents, feeding deterrents and signalling molecules. Metabolomic studies have distinguished metabolic hubs and pathways that support stress responses, providing insight into the exact molecular foundation of maize defense systems [17].
Metabolic adaptations
Chemical defense: With an expansive array of secondary metabolites, comprising benzoxazinoids, terpenoids and phenolics, maize uses its chemical defenses not only against herbivores but also against pathogens. DIMBOA, which is a benzoxazinoid 2,4-dihydroxy-7-methoxy-1,4-benzoxazin-3-one, is among the most potent deterrents of insect herbivores and suppressive agents of soil-borne pathogens. For example, volatile terpenes and nonvolatile diterpenoid defense compounds, both of which deter feeders and hinder fungal growth, are terpenoids that have insecticidal and antifungal activities. Flavonoids and lignin are phenolic compounds that contribute to maize resistance against fungal pathogens by solidifying the cell wall through the expression of defense-related genes.
Indirect defense: Maize indirectly defends itself by releasing Volatile Compounds (VOCs) and sending signals underground that enable it to attract natural enemies of herbivorous and disease-causing organisms. During herbivore attack, some Volatile Organic Compounds (VOCs), such as terpenes, Green Leaf Volatiles (GLVs) and aromatic compounds, are released. These VOCs act as attack cues that attract parasitoids and predators of herbivores. In addition to belowground communication, the emission of root exudates enriched with allelochemicals and signalling substances attracts beneficial soil organisms, such as mycorrhizal fungi and rhizobacteria, which grow plants and create resistance against soil-borne pathogens.
Integrating multi omics data: Painting a holistic picture: These breakthroughs and innovations in high-throughput omics technologies have also revolutionized our understanding of the complexities of biological systems at unparalleled scales. The genome, transcriptome, proteome, metabolome and epigenome are the core components that provide complementary layers with insights into the structure, function and regulation of biological processes. Combining multiomics data permits the construction of comprehensive models that reflect the structure and time dependence of biotic systems and may reveal answers to unique biological problems while allowing their translational application. In our review, we discuss the methods, difficulties and practical use of integrating multiomics data to compile a complete picture of biological systems.
Data integration frameworks: Sharing several omics datasets requires computational frameworks based on data harmonization, batch effect removal and biological system evaluation through interactions across omics layers. Inside this data integration heading, different approaches can be distinguished: Correlation based, network-based and machine learning methods that imply the utilization of statistical inference, graph theory and dimensionality reduction techniques to discover hidden patterns and linkages in multiomics data. Multidimensional integrative processes, such as integrative Multiomics Analysis (iMOFA), mixOmics and Multiomics Factor Analysis (MOFA), connect and apply multiple omics datasets to identify the biological drivers and regulatory mechanisms underlying diseases.
Network-based approaches capitalize on graph theory and systems biology mannerisms by depicting complex networks consisting of gene regulatory networks, protein protein interaction networks and metabolic networks from multiomics data. Network analysis highlights important nodes, modules and pathways that primarily drive cell processes and features, revealing the purpose of biological phenomena and disease mechanisms at the molecular level. Pathway enrichment analysis involves the integration of omics data with curated pathway databases such as KEGG and GO, which enables the detection of pathways and biological processes involved in disease states [18].
The blending of multiomics data conceptualizes the turning to a novel method in biology. A scientist with this incredible ability is able to discover the complex structure of living creatures and she is in a position to find fundamental answers from the fields of biology, medicine and agriculture. The workshop is an opportunity for which the ideas, barricades and prospectus of multiomics integration can be evaluated. Furthermore, the overall effects of both scientific progress and therapy will also be assessed.
Insights from multiomics integration: Comprehensive understanding of biological systems: Multiomics fills in the gaps between the gene, expression, protein, metabolism and epigenetic layers and thus, it will help in personalized medicine and become a catalyst for humanizing biological comprehension. Researchers can use different approaches, such as genomics, proteomics and metabolomics, to integrate omics datasets for the purpose of finding hidden connectivity, revealing new biomarkers and disclosing molecular pathways in diseases though the optimum combination of different methods.
Identification of regulatory networks and pathways: The use of a multimodal dataset will create space for the detection of regulatory networks, signalling pathways and metabolic pathways, which in most cases determine disease states. Genomics, transcriptomics and epigenomics are usually the basis for numerous research directions. The obtained data can thus be applied in the construction of gene regulatory networks, identification of transcription factor-binding sites and landscape visualization of chromatin accessibility. Such strategies will assist in delineating gene expression rules and epigenetic manipulations.
Precision medicine and personalized healthcare: For multifactor analytics, which ultimately leads to precision medicine, multifactor data analysis helps introduce interventions that take into account the molecular biorhizomes of each individual. The application of genomic, transcriptomic, proteomic and metabolomic biomarker screening of patient samples can help physicians subclassify patients, identify therapeutic responses and identify personal treatments with fewer side effects for each person individually.
Challenges in multiomics integration: Data integration and harmonization: Similar to datasets with various omics methods, such as whole-genome sequencing, metabolomic analysis, proteomic profiling, normalization, standardization and integration are difficulties. The multiomics approach involves many methods and various platforms and addresses the issue of variability in data as well as problems with batch effects, data quality and the difficulty of reproducing the research.
Computational complexity and interpretability: Analysing multiomics data requires intricate computer infrastructure and a variety of algorithms that only experienced bioinformaticians, statisticians and systems biologists can use. The process of interpreting integrated omics results together with the complexity of data visualization, the development of new hypotheses and biological interpretation are the main obstacles in the field [19].
Biological and clinical validation: The application of multiomics in both the clinical and agricultural arena is dependent on meticulous validation in separate cohorts, experimental models and functional expression systems. Blending multiomics data supplemented with clinical results, phenotypic traits and experimental validations is important for identifying causal relationships, the functionality of biomarkers in the actual world setting and the efficacy of therapeutics.
Emerging technologies and platforms: The employment of next-generation omics technologies, single-cell sequencing techniques, spatial omics, and multimodal imaging would push the boundaries in multiomics integration, which are capable of analysing cellular heterogeneity, spatial organization and dynamic complexity.
Systems biology and computational modelling: The conjunction of multiomics data with biological computational models, including dynamic models, network models and machine learning methods, will provide an enhanced understanding of biological processes, predictive modelling of disease growth and rational design of therapeutic interventions.
Data sharing and collaborative research: Dedicated multiomics data repositories, consortia and community resources will empower the sharing of data, cooperation and replicability in multiomics research. Over the years, open data platforms, metadata standards and data-sharing agreements will provide enough support for interdisciplinary collaboration and consequently lead to breakthroughs in science.
In summary, multiomics integration has emerged as a unique tool that allows further understanding of biological complexity, which, in turn, will drive innovation in research, medicine and agriculture. Tackling the merits and demerits of multiomics platform integration implies its proper application for understanding health and disease, quick medicine discovery and development for further application in personalized and precision farming [20].
Future perspectives and areas for further research
Single-cell multiomics integration: New biotechnologies that enable single-cell examination provide a chance for a better understanding of cell heterogeneity and dynamics with a resolution that was previously impossible. Integrating single-cell genomics, transcriptomics, proteomics and epigenomics data will help us to identify cell type-specific regulatory networks, signalling pathways that are in action and cellular responses leading to biological outcomes caused by the environment.
Temporal dynamics of multiomics profiles: The patterns of temporal dynamics across multiomics profiles at multiple time points or under different experimental conditions will reveal temporal expression, changing molecular associations and responses to stimuli. By performing multiomics studies under longitudinal conditions, clinicians and researchers can monitor the progression of the disease, response to therapy and recovery, which, in turn, can be exploited for more tailored intervention strategies.
Multiomics data integration in systems biology models: The merger of multiomics information with in silico systems (e.g., genome-scale metabolic models (GEMs and Boolean networks will improve our capacity to forecast the cellular phenotypes and gene regulatory networks and the interactions of these networks. Systems biology refers to the application of computational science to simulate and analyse intricate biochemist processes, creating a supportive environment for hypothesis building and deductive experiments.
Multiomics atlas and data repositories: Creating an all embracing multiomics atlas and shared data depositories will open the way to data exchange, joint research and meta-analysis for the multiomics community. Centralized resource portfolios, i.e., HCA (Human Cell Atlas and TCGA (Cancer Genome Atlas, furnish data about very important tissue-specific gene expression profiles, large regulatory networks and disease signatures.
Multiomics integration in precision agriculture: Multiple omics integration into plant breeding, crop protection and agricultural sustainability will have greater force. Blending of genomics, transcriptomics, metabolomics and environment data allows for the determination of genetic markers, metabolic pathways and physiological traits directly related towards agronomic traits, stress tolerance and yields potential.
To create maize that is innately highly durable against fungal pathogens, we need to implement multiple approaches. Hence, by employing conventional breeding techniques along with state of-the-art technologies such as MAS, genome editing and speed breeding, it is possible to generate maize varieties with remarkable and broad-spectrum immunity. Constant research and poisoning fortifications to avoid future impediments are key to guaranteeing food safety and reducing the harvest losses caused by fungal diseases. The recognition and genetic use of resistance genes and mutations, which often point in this direction, are some of the factors that are used in modern plant breeding systems to improve the immunity of crops to pathogenic microorganisms. Researchers are now able to analyse genetic variations, genotypes and gene control mechanisms to further their understanding of the core genetic structure of immunity and learn ways to develop naturally induced mutations for permanent sustainable agriculture and worldwide food security. Genome-Wide Association Studies (GWASs) have been successfully brought to the forefront as advanced methods for scrutinizing and revealing the genetic components underlying the variations in complex traits and diseases associated with different organisms. By reducing economic costs, geneticists and epidemiologists have been able to rely on existing data and natural genetic differences as their sources to determine the variations underlying phenotypic variation, thus improving the accuracy of animal breeding, precision medicine and basic biological research. On account of growing sophistication in the methodology and tools of GWASs, it has become possible for experts to be able to determine causal factors and outcome factors faster. Gene expression alterations are a component of cell stress adaptation and range from simple to complex living systems. Researchers have applied molecular, genomic and computational technologies that help deduce the regulatory mechanisms, functional effects and adaptive tactics underlying stress responses. Tailored approaches that use single cell transcriptomics, spatial transcriptomics and multiomics integration are used to help crack the code of the movement and activity of signalling pathways at the cellular and tissue levels in the case of an infection. Efforts to manipulate crucial pathways and nodes for the regulation of the immune response and immune system activation seem to be the most promising. Precision medicine techniques based on a close understanding of host-interactions and regulatory networks can be identified as a new way to develop immunomodulatory therapies and vaccines against infectious diseases.
All the data described in this research paper are presented in tables and figures because they are real time data.
No conflicting interests are disclosed by the author.
For this research, no funding was obtained.
All the efforts ware carried out by the author Muhammad Amir Sial & Muhammad Aoun Munir; in this paper no other author was involved.
Authors have read, understood and complied with the statement on "Ethical responsibilities of authors", as found in the instructions for authors.
Citation: Sial MA, Munir MA (2025) Investigating the Effect of Fungal Pathogens on Maize Growth and Omics to Cope with Pathogenic Effects. J Plant Pathol Microbiol. 16:753.
Copyright: © 2025 Sial MA, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.