Appearance
This report is written by MaltSci based on the latest literature and research findings
What is the role of genetic mutations in disease?
Abstract
The understanding of genetic mutations has fundamentally transformed biomedical research, revealing their critical role in the etiology of various diseases. Genetic mutations, defined as alterations in the DNA sequence, can manifest in numerous forms including point mutations, insertions, deletions, and chromosomal alterations, each significantly influencing disease susceptibility and progression. For example, germline mutations are responsible for hereditary conditions such as cystic fibrosis, while somatic mutations are often implicated in cancer development. Recent advancements in next-generation sequencing technologies have enhanced our ability to identify and characterize these mutations, providing invaluable insights into their functional consequences. This review systematically categorizes genetic mutations, explores the mechanisms by which they induce disease, and emphasizes the role of environmental factors in modulating their effects. Additionally, the review discusses the implications of genetic testing and the transformative potential of precision medicine in clinical practice. The insights gained from this exploration underscore the necessity for continued research into the complexities of genetic influences on health, aiming to foster a more nuanced understanding of human disease and to advance therapeutic strategies.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Types of Genetic Mutations
- 2.1 Point Mutations
- 2.2 Insertions and Deletions
- 2.3 Copy Number Variations
- 2.4 Chromosomal Alterations
- 3 Mechanisms of Mutation-Induced Disease
- 3.1 Oncogenesis
- 3.2 Monogenic Disorders
- 3.3 Multifactorial Diseases
- 4 Role of Environmental Factors
- 4.1 Carcinogens and Mutagens
- 4.2 Lifestyle Influences
- 4.3 Epigenetic Modifications
- 5 Advances in Genetic Testing and Implications
- 5.1 Next-Generation Sequencing
- 5.2 Clinical Applications
- 5.3 Ethical Considerations
- 6 Future Directions in Research
- 6.1 Gene Therapy
- 6.2 Precision Medicine
- 6.3 Ongoing Clinical Trials
- 7 Summary
1 Introduction
The understanding of genetic mutations has transformed the landscape of biomedical research, revealing their fundamental role in the etiology of various diseases. Genetic mutations, defined as alterations in the DNA sequence, can manifest in numerous forms, including point mutations, insertions, deletions, and chromosomal alterations. These variations not only contribute to the phenotypic diversity among individuals but also significantly influence disease susceptibility, progression, and severity. For instance, germline mutations can lead to hereditary conditions such as cystic fibrosis or sickle cell anemia, while somatic mutations are often implicated in cancer development and progression [1][2]. As such, a comprehensive understanding of genetic mutations is critical for advancing diagnostic and therapeutic strategies in clinical practice.
The significance of genetic mutations extends beyond mere disease causation; they are pivotal in shaping our understanding of complex biological processes and the evolution of species. Recent advancements in next-generation sequencing technologies have greatly enhanced our ability to identify and characterize these mutations, thereby providing invaluable insights into their functional consequences [3]. Furthermore, the interplay between genetic and environmental factors complicates the landscape of disease etiology. Acquired mutations can arise from environmental exposures, lifestyle choices, and epigenetic modifications, highlighting the multifaceted nature of genetic influences on health [4][5].
Despite the wealth of knowledge accumulated over the past few decades, significant gaps remain in our understanding of the precise mechanisms by which genetic mutations contribute to various diseases. The current research landscape is characterized by a growing emphasis on elucidating the molecular pathways affected by mutations and their interactions with environmental factors. For instance, studies have shown that specific mutations can influence the risk of developing multifactorial diseases such as diabetes and cardiovascular conditions, which are influenced by both genetic predispositions and lifestyle factors [6][7]. Additionally, the recognition of de novo mutations as critical contributors to neurodevelopmental disorders underscores the need for continued exploration of mutation-induced disease mechanisms [8].
This review is organized into several key sections that systematically address the multifaceted role of genetic mutations in disease. The second section will categorize the various types of genetic mutations, including point mutations, insertions and deletions, copy number variations, and chromosomal alterations, providing a foundational understanding of their distinct implications for health. The third section will delve into the mechanisms by which these mutations induce disease, focusing on oncogenesis, monogenic disorders, and multifactorial diseases. Following this, the fourth section will explore the role of environmental factors in modulating the effects of genetic mutations, emphasizing the interactions between carcinogens, lifestyle influences, and epigenetic modifications.
Advancements in genetic testing and their clinical implications will be discussed in the fifth section, highlighting the transformative potential of next-generation sequencing in personalized medicine. Ethical considerations surrounding genetic testing will also be addressed, as the implications of genetic knowledge continue to evolve. Finally, the review will conclude with a discussion of future research directions, including gene therapy, precision medicine, and ongoing clinical trials, which aim to harness our understanding of genetic mutations for improved health outcomes.
In summary, the intricate relationship between genetic mutations and disease underscores the importance of continued research in this field. By synthesizing current knowledge and exploring the complexities of genetic influences on health, this review aims to provide a clearer understanding of the significance of genetic mutations in biomedical research and clinical practice. The insights gained from this exploration are not only crucial for advancing therapeutic strategies but also for fostering a more nuanced understanding of human health and disease.
2 Types of Genetic Mutations
2.1 Point Mutations
Genetic mutations play a significant role in the pathogenesis of various diseases, with point mutations being a prominent type of mutation associated with a wide range of genetic disorders. Point mutations are alterations in a single nucleotide base pair in the DNA sequence, which can lead to significant changes in gene function and protein synthesis.
Point mutations can be classified into different categories based on their effects on protein coding. They may lead to missense mutations, where a single amino acid in a protein is changed, potentially altering its function. For instance, point mutations affecting genes coding for tRNAs or mitochondrial respiratory chain proteins are inherited maternally and are associated with a variety of clinical manifestations, including mitochondrial cytopathies [9]. Additionally, mutations in critical driver genes such as TP53, BRCA1, BRCA2, EGFR, KRAS, and BRAF are essential for cancer risk assessment and treatment response prediction [10].
Moreover, point mutations are implicated in both hereditary and sporadic diseases. For example, de novo germline point mutations have been identified as significant contributors to both rare and common genetic disorders, including neurodevelopmental diseases such as autism and schizophrenia [8]. The mutational target size influences the frequency of diseases caused by these mutations, and factors such as advanced paternal age are associated with an increased risk of de novo mutations, which can lead to congenital anomalies [11].
Point mutations can also affect splicing patterns, leading to altered mRNA processing and functional consequences for the resulting proteins. Studies indicate that a substantial proportion of point mutations can disrupt exonic splicing enhancers (ESEs), resulting in exon skipping, which can have severe implications for protein function [12]. Furthermore, point mutations at splice junctions have been shown to account for a significant number of genetic diseases, suggesting that alterations in splicing patterns may be a frequent consequence of these mutations [13].
The frequency and impact of point mutations can vary widely, and their study has been greatly enhanced by advancements in sequencing technologies. Whole-exome and whole-genome sequencing have allowed for a more comprehensive understanding of the role of point mutations in various diseases, revealing that while individually rare, they can collectively account for a significant portion of heritability in complex genetic diseases [8].
In summary, point mutations serve as critical elements in the etiology of genetic diseases, influencing not only the structure and function of proteins but also the splicing and regulation of gene expression. Their implications are profound, ranging from inherited disorders to acquired mutations in cancers, thereby highlighting the necessity for continued research into their mechanisms and effects on human health.
2.2 Insertions and Deletions
Genetic mutations play a significant role in the etiology of various diseases, with insertions and deletions (indels) being a critical type of mutation that contributes to genetic variability and disease pathogenesis. Indels are particularly noteworthy as they can lead to significant alterations in the coding sequences of genes, potentially resulting in functional consequences that may manifest as genetic disorders.
Insertions and deletions are known to constitute up to 25% of genomic variants in humans and are involved in complex evolutionary processes, including genomic rearrangements, adaptation, and speciation (Redelings et al., 2024). They are also a substantial source of natural genomic variation and can lead to pronounced evolutionary differences among genomes. For instance, research indicates that indels can cause numerous genetic diseases, highlighting their impact on human health (Kvikstad et al., 2007).
In the context of disease, specific studies have analyzed the contribution of indels to genetic mutations. For example, a study by Darvasi and Kerem (1995) found that 21% of disease-causing mutations identified in the coding regions of three human genes (cystic fibrosis transmembrane conductance regulator, beta globin, and factor IX) were insertion or deletion mutations of 4 base pairs or less. Notably, these mutations often occurred within repetitive sequences, classified as slipped-strand mispairing (SSM) mutations, which were found to contribute significantly to the generation of indels in human genes.
Furthermore, the mechanisms underlying the occurrence of insertions and deletions have been explored. Kvikstad et al. (2007) demonstrated that both replication and recombination are significantly associated with the occurrence of small indels. Their findings suggest that insertions are more strongly associated with factors linked to recombination, while deletions are predominantly associated with replication-related features. This distinction is crucial, as it indicates that the mechanisms guiding these two types of mutations may differ, leading to varied impacts on gene function and, consequently, on disease phenotypes.
The implications of indels extend beyond mere genetic variability; they can lead to functional disruptions in genes, resulting in a spectrum of genetic diseases. For instance, when an indel occurs within a coding region, it may lead to frameshift mutations, which can produce truncated proteins or alter protein function, thereby contributing to disease pathology. The identification and understanding of these mutations are vital for elucidating the mechanisms of disease, developing diagnostic tools, and informing therapeutic strategies.
In summary, insertions and deletions are a significant source of genetic mutations that can profoundly impact human health. Their role in generating genetic diversity and their association with various diseases underscore the importance of studying these mutations in the context of genetics and medicine. Understanding the mechanisms behind indels and their contributions to disease can provide valuable insights into genetic disorders and inform future research and clinical practices.
2.3 Copy Number Variations
Copy number variations (CNVs) are a significant form of genetic mutation characterized by alterations in the number of copies of specific segments of DNA. These variations can manifest as deletions or duplications of chromosomal segments and play a crucial role in various diseases, including neuropsychiatric disorders, cardiovascular diseases, and metabolic disorders.
The role of CNVs in disease is multifaceted. They can lead to changes in gene dosage, which in turn affects gene expression and contributes to phenotypic diversity and disease susceptibility. For instance, in neuropsychiatric conditions such as autism and schizophrenia, CNVs have been implicated in the genetic underpinnings of these disorders. Research has shown that when CNVs alter the dosage of genes critical for normal brain development and functioning, they may result in severe disorders. Numerous loci associated with these conditions have been identified, providing valuable insights for neuropsychiatric research (Cook & Scherer 2008) [14].
Moreover, CNVs are not limited to neuropsychiatric diseases. They have been recognized as important contributors to cardiovascular disorders as well. For example, a review highlighted that CNVs may influence lipoprotein and metabolic phenotypes, suggesting their potential role in the etiology of conditions like familial hypercholesterolemia, where CNVs in the low-density lipoprotein receptor gene have been found to be causative (Pollex & Hegele 2007) [15].
The complexity of CNVs extends to their involvement in common diseases. Recent genome-wide studies have demonstrated that CNVs are prevalent in the general population and can be associated with various phenotypes, particularly those related to immune response and environmental factors (Girirajan et al. 2011) [16]. This indicates that CNVs not only contribute to rare Mendelian disorders but also to the genetic architecture of complex diseases, thereby underscoring their importance in understanding the inherited basis of human variation and disease.
In addition to their role in disease, CNVs also provide insights into evolutionary processes. The presence of CNVs in populations contributes to genetic diversity and can influence susceptibility to diseases through structural polymorphisms, which vary in frequency across different populations (McCarroll & Altshuler 2007) [17]. This variation can create different chromosomal architectures that may either predispose individuals to disease or confer protection against certain conditions.
In summary, CNVs represent a significant class of genetic mutations with profound implications for disease susceptibility and phenotypic variation. Their study has the potential to elucidate the genetic basis of both rare and common diseases, enhance our understanding of human genetics, and pave the way for improved diagnostic and therapeutic strategies.
2.4 Chromosomal Alterations
Genetic mutations play a critical role in the etiology of various diseases, including both inherited and acquired conditions. Mutations can be classified into two main types: gene mutations, which affect single genes, and chromosomal mutations, which involve larger segments of DNA that may contain multiple genes or entire chromosomes. Chromosomal alterations can lead to significant changes in genetic material and are implicated in a range of diseases, particularly cancers.
Chromosomal mutations can result in structural changes such as deletions, duplications, inversions, or translocations of chromosomal segments. These alterations can disrupt normal gene function and regulation, leading to disease. For example, the Philadelphia chromosome, a well-known chromosomal aberration, is formed by a reciprocal translocation between chromosomes 9 and 22, resulting in the BCR-ABL1 fusion gene, which acts as a potent oncogene in chronic myeloid leukemia (CML) (Rumpold & Webersinke 2011). This mutation is critical for the diagnosis and treatment of CML, as it drives the pathogenesis of the disease and influences therapeutic strategies.
Moreover, chromosomal mutations can also be associated with an increased risk of various diseases. Studies indicate that chromosomal abnormalities are significant prognostic factors in hematological malignancies, influencing treatment outcomes and survival rates (Schilling et al. 2005). The presence of specific genetic changes can lead to risk-adapted therapeutic approaches, demonstrating the importance of understanding chromosomal alterations in medical practice.
In addition to their role in cancer, chromosomal mutations can also contribute to other inherited genetic disorders. The mutation rates for inherited chromosomal alterations are age-dependent and can be influenced by environmental factors, including exposure to mutagens. Research has shown that exposure to physical or chemical mutagens can increase the incidence of both gene and chromosomal mutations (Evans 1988).
Furthermore, the implications of chromosomal mutations extend beyond the direct consequences of the mutations themselves. The accumulation of such mutations can lead to somatic mosaicism, where different cells within the same individual have different genetic makeups. This phenomenon has been observed in aging populations and is associated with various age-related diseases, including cancer and cardiovascular conditions (Walsh et al. 2022).
In summary, chromosomal alterations are a significant category of genetic mutations that contribute to the pathogenesis of various diseases, particularly cancers and inherited disorders. Understanding these mutations is crucial for developing targeted therapies and improving patient outcomes in clinical settings. The ongoing research into the mechanisms and consequences of chromosomal mutations continues to provide valuable insights into their role in human health and disease.
3 Mechanisms of Mutation-Induced Disease
3.1 Oncogenesis
Genetic mutations play a critical role in the development of diseases, particularly cancer, by disrupting normal cellular functions and promoting oncogenesis. The initiation and progression of cancer are often attributed to a series of genetic alterations that affect key regulatory genes involved in cellular processes such as proliferation, differentiation, and DNA repair.
One of the fundamental mechanisms by which genetic mutations contribute to oncogenesis is through the inactivation of tumor suppressor genes and the activation of oncogenes. Tumor suppressor genes typically function to regulate cell growth and maintain genomic stability. When these genes are mutated, their ability to suppress uncontrolled cell division is compromised, leading to tumorigenesis. Conversely, oncogenes, which promote cell growth and division, can become hyperactivated due to mutations, further driving the malignant transformation of cells (Dixon and Kopras, 2004) [18].
Moreover, chromosomal translocations and other genetic rearrangements can lead to the formation of fusion genes that have oncogenic properties. These alterations can occur sporadically or may be inherited, significantly increasing the risk of cancer, particularly in childhood cancers where specific genetic alterations are often used for diagnosis and prognosis (Ganjavi and Malkin, 2002) [19].
The mutational landscape of cancer is also influenced by epigenetic changes, which can occur alongside genetic mutations. Epigenetic modifications, such as DNA methylation and histone modifications, can alter gene expression without changing the underlying DNA sequence. These changes can contribute to the development of cancer by silencing tumor suppressor genes or activating oncogenes, thereby linking environmental factors and genetic predispositions to cancer development (Stadler and Allis, 2012) [20].
Recent studies have indicated that mutations are not randomly distributed across the genome but are significantly influenced by intrinsic molecular processes, such as transcription and DNA repair mechanisms. This interplay can create a characteristic mutational landscape in tumors, affecting their behavior and response to treatment (Lim et al., 2017) [5].
Furthermore, the concept of "oncogenic competence" highlights that the context in which mutations occur—shaped by cellular lineage, differentiation state, and the microenvironment—plays a crucial role in determining whether a mutation will lead to malignant transformation (Pavinato and Baggiolini, 2025) [21].
In summary, genetic mutations are central to the oncogenic process, as they disrupt normal cellular regulation and contribute to the hallmarks of cancer. The interplay between genetic and epigenetic factors, as well as the cellular context in which these mutations occur, underscores the complexity of cancer biology and the necessity for targeted therapeutic strategies that consider these dynamics.
3.2 Monogenic Disorders
Genetic mutations play a pivotal role in the etiology of various diseases, particularly monogenic disorders, which are caused by mutations in a single gene. These mutations can lead to a wide array of phenotypic expressions and clinical manifestations due to their direct impact on protein function and cellular pathways.
Monogenic disorders are characterized by Mendelian inheritance patterns, where mutations can be dominant, recessive, or X-linked. They are often associated with significant alterations in the structure or function of proteins, which can disrupt normal biological processes. For instance, mutations in genes such as alpha-synuclein or LRRK2, which are implicated in Parkinson's disease, can lead to neurodegeneration either through a toxic gain of function or a loss of protective functions in neuronal cells (Gasser 2009) [22]. Similarly, mutations in genes associated with systemic lupus erythematosus (SLE) can cause dysregulation of immune pathways, resulting in autoimmune manifestations such as kidney and skin issues (Jia et al. 2023) [23].
The identification of disease-associated genes has been significantly advanced by technologies such as next-generation sequencing, allowing for the genetic characterization of previously undiagnosed patients. This technological progress has revealed a complex landscape of mutations that can contribute to disease phenotypes. For example, in monogenic lupus, over thirty genetic variations have been identified, highlighting how single-gene mutations can lead to immune dysfunction characterized by the activation of innate immune cells and the production of pro-inflammatory cytokines (Jia et al. 2023) [23].
Moreover, genetic mutations can also have implications beyond monogenic disorders. They can influence the risk of developing multifactorial diseases, as seen in stroke, where both genetic predispositions and environmental factors interplay (Rubattu et al. 2000) [24]. The genetic architecture of complex disorders often includes rare monogenic variants that contribute to disease susceptibility, underscoring the importance of understanding these genetic factors for developing targeted therapies.
In terms of therapeutic strategies, gene therapy has emerged as a promising approach for addressing monogenic disorders. Advances in gene editing technologies, such as CRISPR/Cas, have opened new avenues for correcting mutations at the genomic level, potentially reversing the pathological consequences of genetic defects (Zhang and Wu 2024) [25]. However, challenges remain in terms of efficient delivery, expression stability, and minimizing immunogenic responses.
In conclusion, genetic mutations serve as the fundamental basis for monogenic disorders, directly influencing the pathophysiology of these diseases through alterations in protein function and cellular mechanisms. The ongoing exploration of genetic mutations not only enhances our understanding of disease mechanisms but also informs the development of innovative therapeutic strategies aimed at correcting these genetic defects.
3.3 Multifactorial Diseases
Genetic mutations play a significant role in the etiology of multifactorial diseases, which are complex conditions influenced by both genetic and environmental factors. Unlike monogenic diseases caused by single gene mutations, multifactorial diseases, such as cardiovascular diseases, cancers, and diabetes, are typically associated with variations in multiple genes (polymorphisms) that each have a modest effect on disease susceptibility at the individual level. However, these genetic variations are prevalent in the population and can collectively contribute to a high attributable risk for these diseases (Tiret 2002) [26].
The interaction between genetic mutations and environmental factors is critical in the manifestation of multifactorial diseases. Genetic factors may act as response modifiers, influencing how individuals respond to various environmental triggers such as stress, diet, and exposure to toxins. This interaction can amplify the phenotypic expression of susceptibility genes, meaning that the presence of certain environmental factors can significantly heighten the risk of developing a disease in genetically predisposed individuals (Tiret 2002) [26].
Moreover, gene therapy has emerged as a potential strategy for treating diseases associated with genetic mutations, particularly in diabetes, which is a polygenic and multifactorial condition. The ability to alter genetic heritage and potentially prevent or treat diseases linked to specific mutations has garnered significant interest, although ethical considerations and the complexities of multifactorial diseases complicate this field (Bertolaso et al. 2010) [27].
Recent advances in genomic technologies, such as high-throughput RNA sequencing, have revealed that non-coding RNAs, particularly long non-coding RNAs (lncRNAs), are also involved in the pathogenesis of multifactorial diseases. These lncRNAs can be deregulated in disease states and may serve as important biomarkers for diagnosis and therapeutic targets (Cipolla et al. 2018) [28].
Furthermore, neurodegenerative diseases, including Alzheimer’s and Parkinson’s diseases, exemplify the complex interplay of genetic mutations and environmental factors. Research indicates that neuroinflammation, potentially exacerbated by genetic predispositions, is a common mechanism linking these diseases to environmental risk factors (Boyd et al. 2022) [29]. This suggests that genetic variations may predispose individuals to heightened inflammatory responses, thereby increasing their risk for neurodegeneration when exposed to environmental insults.
In summary, genetic mutations are integral to the development of multifactorial diseases, influencing individual susceptibility and disease progression through complex interactions with environmental factors. Understanding these interactions is crucial for advancing therapeutic strategies and improving disease management.
4 Role of Environmental Factors
4.1 Carcinogens and Mutagens
Genetic mutations play a significant role in the etiology of various diseases, particularly in the context of environmental factors such as carcinogens and mutagens. De novo mutations, which are new mutations that arise in an individual's genome, contribute substantially to sporadic psychiatric and developmental disorders. A recent study highlighted that environmental carcinogens can act as drivers of these causal de novo mutations in neurodevelopmental disorders, suggesting a critical interplay between genetic susceptibility and environmental exposures [30].
Carcinogenesis is inherently multifactorial, involving both genetic and environmental influences. It is increasingly recognized that virtually all cancer cases involve a combination of inherited genetic predispositions and environmental factors, which include chemical exposure, ionizing radiation, and oncogenic viruses. The process of carcinogenesis consists of several stages: initiation, which involves irreversible changes to the genome; promotion, which is associated with prolonged exposure to carcinogens; and progression, where further genetic changes lead to tumor growth and metastasis [31].
Specific environmental agents, including polycyclic aromatic hydrocarbons and radiation, have been shown to disproportionately affect genes associated with neurodevelopmental disorders, such as autism spectrum disorders and schizophrenia. In this context, longer gene sequences were found to be more susceptible to mutations, while increased gene expression correlated with lower mutation rates in coding sequences [30]. This indicates that the characteristics of the genes themselves, alongside the environmental context, are crucial in determining mutation rates.
Moreover, the National Institute of Environmental Health Sciences has conducted extensive research demonstrating that exposure to mutagenic chemicals can lead to new mutations in mammalian germ cells, thereby contributing to genetic diseases in the human population [32]. The mutagenic potential of environmental chemicals is supported by animal model studies, reinforcing the idea that these exposures can lead to significant genetic alterations.
In addition to direct mutations, epigenetic modifications—heritable changes that do not alter the DNA sequence—also play a crucial role in disease development. Environmental and occupational exposures can lead to epigenetic alterations that influence gene expression and contribute to the pathogenesis of diseases, including cancer [33]. This interplay between genetic and epigenetic factors underscores the complexity of disease etiology, particularly in relation to environmental influences.
The impact of environmental factors on genetic susceptibility is particularly pronounced in children, who may be more vulnerable to the effects of such exposures during critical periods of development. Genetic variations can influence how individuals metabolize and respond to environmental agents, further complicating the relationship between genetics and environmental exposures [34].
In summary, genetic mutations are integral to the development of various diseases, with environmental factors acting as significant contributors to the mutation process. The interaction between genetic predispositions and environmental exposures, particularly from carcinogens and mutagens, is essential for understanding the multifaceted nature of disease etiology.
4.2 Lifestyle Influences
Genetic mutations play a critical role in the etiology of various diseases, influencing both Mendelian and complex conditions. These mutations can be categorized into germline mutations, which are inherited and present in the reproductive cells, and somatic mutations, which occur in non-reproductive cells and are often associated with cancer.
Germline mutations are the principal cause of heritable diseases and contribute to the genetic diversity observed in human populations. They can lead to conditions such as Mendelian diseases, where a single gene mutation has a direct impact on disease phenotype. For example, mutations in genes like BRCA1 and BRCA2 are well-documented to significantly increase the risk of breast and ovarian cancers, demonstrating how specific genetic alterations can predispose individuals to certain health outcomes (Muggia et al. 2011) [35].
In contrast, somatic mutations are typically acquired during a person's lifetime and are prevalent in cancer development. These mutations can arise from various factors, including environmental exposures and intrinsic cellular processes. For instance, somatic mutations in driver genes can lead to clonal expansion in tissues, contributing to tumorigenesis (Walsh et al. 2022) [36]. Furthermore, recent studies have indicated that the distribution of mutations is non-random and is influenced by intrinsic molecular processes such as DNA replication and repair mechanisms (Lim et al. 2017) [5].
The impact of genetic variations extends beyond single-gene disorders. Complex diseases, such as diabetes and heart disease, are influenced by multiple genetic factors interacting with environmental elements. Genetic variations can affect susceptibility to these diseases, influencing both the initiation and progression of disease states (Kamatani and Nakamura 2021) [3]. For instance, variations in cytokine genes have been linked to inflammatory responses that are central to chronic diseases (Duff 2006) [6].
Additionally, genetic mutations can also affect drug efficacy and adverse reactions, which is particularly relevant in the context of pharmacogenomics. Understanding the genetic basis of drug response can lead to more personalized medicine approaches, tailoring treatments based on an individual's genetic makeup (Strynatka et al. 2018) [37].
Overall, genetic mutations are pivotal in shaping disease susceptibility and progression. They not only provide insight into the mechanisms of disease but also offer potential avenues for therapeutic interventions and personalized treatment strategies. Understanding these genetic underpinnings, in conjunction with environmental factors, is essential for advancing medical research and improving health outcomes.
4.3 Epigenetic Modifications
Genetic mutations play a significant role in the etiology of various diseases, influencing not only the risk of developing certain conditions but also the severity and progression of these diseases. Traditional views in genetics focused on direct alterations in the DNA sequence as the primary cause of diseases. However, the contemporary understanding has expanded to include the interplay between genetic factors and epigenetic modifications, which can modulate gene expression without altering the underlying DNA sequence.
Epigenetic modifications, such as DNA methylation and histone modifications, are critical in regulating gene expression and can be influenced by environmental factors. These modifications serve as a bridge between genetic predispositions and environmental exposures, thus shaping the phenotypic outcomes in individuals. For instance, environmental factors such as diet, stress, and toxins can induce epigenetic changes that may lead to disease development or progression, thereby complicating the relationship between genetic mutations and disease manifestation [38].
Research indicates that epigenetic alterations can impact various diseases, including cancer, neurodegenerative disorders, and metabolic syndromes. In cancer, for example, aberrant epigenetic signaling is increasingly recognized as a central component of tumorigenesis, with epigenetic changes contributing to the activation of oncogenes or silencing of tumor suppressor genes [39]. Moreover, studies have shown that the epigenetic landscape can be reshaped by environmental factors, which further complicates the pathogenesis of diseases [40].
The interplay between genetic mutations and epigenetic modifications is particularly relevant in complex diseases, where both genetic susceptibility and environmental influences converge. Genetic variability can determine an individual's epigenetic profile, which may then be further modified by environmental exposures. This dynamic interaction is crucial for understanding disease etiology and for developing targeted therapies. For example, the identification of epigenetic modifiers as therapeutic targets has shown promise in treating various conditions, including cancer and neurodegenerative diseases [41].
Overall, the relationship between genetic mutations, environmental factors, and epigenetic modifications highlights the complexity of disease etiology. Understanding these interactions is essential for identifying individuals at risk and developing effective therapeutic strategies, thereby underscoring the importance of integrating genetic, epigenetic, and environmental data in biomedical research [42].
5 Advances in Genetic Testing and Implications
5.1 Next-Generation Sequencing
Genetic mutations play a pivotal role in the pathogenesis of various diseases, serving as critical factors that contribute to disease development, progression, and variability in clinical outcomes. The understanding of genetic mutations has advanced significantly, particularly with the advent of next-generation sequencing (NGS) technologies, which allow for comprehensive analysis of genetic alterations across the genome.
Mutations can be classified into different categories, including single nucleotide polymorphisms (SNPs), insertions, deletions, and copy number variations. These mutations can lead to alterations in protein function, disrupt regulatory elements, and affect gene expression, ultimately influencing an individual's susceptibility to diseases. For instance, mutations in key genes have been implicated in the pathogenesis of various conditions, including cancers, cardiovascular diseases, and neurodegenerative disorders.
In the context of primary myelofibrosis (PMF), a type of myeloproliferative neoplasm, mutations in driver genes such as JAK2, CALR, and MPL have been well characterized. These mutations are crucial for understanding the disease's biological consequences and the heterogeneous clinical presentations observed in patients. Recent findings suggest that the dynamic structure of PMF involves competing disease subclones and sequentially acquired genomic events, which complicate the clinical landscape and highlight the importance of integrating molecular information into prognostic scoring systems for therapeutic decision-making (Rumi et al. 2020) [43].
Moreover, the impact of genetic mutations extends beyond the identification of disease mechanisms; it also informs treatment strategies. For example, the presence of specific mutations can dictate the choice of targeted therapies in cancer treatment, exemplifying the clinical relevance of genetic testing. In the case of chronic myeloid leukemia (CML), the BCR-ABL1 fusion gene is a hallmark of the disease, and its presence guides the use of tyrosine kinase inhibitors. However, in advanced stages of CML, the emergence of additional mutations can lead to treatment resistance, emphasizing the need for ongoing genetic monitoring (Rumpold & Webersinke 2011) [44].
Next-generation sequencing has revolutionized the landscape of genetic testing by enabling high-throughput analysis of multiple genes simultaneously, thereby facilitating the identification of both rare and common genetic variants associated with diseases. This technology enhances our understanding of complex genetic interactions and their implications for disease pathogenesis. For instance, in the realm of migraine, advances in NGS have uncovered various genetic factors that contribute to both monogenic and polygenic forms of the disorder, providing insights into the underlying biological pathways and potential therapeutic targets (Sutherland et al. 2019) [45].
In summary, genetic mutations serve as fundamental components in the etiology of numerous diseases, influencing both the biological mechanisms of disease and the clinical approaches to treatment. The integration of next-generation sequencing into clinical practice is poised to enhance the precision of genetic testing, ultimately leading to improved diagnostic, prognostic, and therapeutic outcomes across a wide array of medical conditions.
5.2 Clinical Applications
Genetic mutations play a pivotal role in the development and manifestation of various diseases, significantly influencing phenotypic variations among individuals. These mutations can be broadly categorized into germline mutations, which are inherited and can predispose individuals to certain diseases, and somatic mutations, which occur in non-germline tissues and are primarily associated with cancers and age-related diseases.
Germline mutations are implicated in both Mendelian disorders and complex diseases, such as diabetes and heart diseases. They serve as a substrate for evolutionary processes but also contribute to disease susceptibility. These mutations can affect drug efficacy and adverse reactions, thus highlighting their importance in personalized medicine. Recent advancements in DNA sequencing technologies enable the identification of these genetic variations, which can lead to tailored medical management strategies for affected individuals [1].
Somatic mutations, on the other hand, are a major driver of cancer and are often acquired during a person's lifetime. These mutations can result from environmental factors, replication errors, or DNA repair failures. The complexity and high frequency of these mutations pose challenges for experimental verification of their functions. However, computational approaches, particularly machine learning, have made significant strides in predicting the functional consequences of mutations, thereby aiding in the understanding of their roles in disease [1].
Moreover, the concept of de novo mutations has gained attention, particularly in the context of neurodevelopmental disorders such as autism and schizophrenia. These mutations can be particularly impactful as they may not be present in the parents, leading to unique disease presentations in the offspring [8].
The clinical implications of understanding genetic mutations are profound. With the rise of personalized medicine, the integration of genetic testing into routine clinical practice allows for more accurate diagnoses and tailored treatment plans. Genetic testing can identify mutations that dictate the risk of developing specific diseases, enabling preemptive healthcare measures [46].
In conclusion, genetic mutations are central to the pathogenesis of various diseases, influencing not only the clinical presentation but also the management and treatment options available to patients. The advancements in genetic testing and the understanding of these mutations are transforming the landscape of clinical medicine, paving the way for more effective and personalized therapeutic interventions [3][47].
5.3 Ethical Considerations
Genetic mutations play a pivotal role in the etiology of various diseases, influencing both Mendelian and complex conditions. These mutations can be categorized into germline mutations, which are inherited and contribute to heritable diseases, and somatic mutations, which occur in non-germline cells and are primarily associated with cancers and age-related diseases.
Germline mutations can lead to a range of genetic disorders, including Mendelian diseases characterized by single-gene defects. For instance, de novo mutations have been implicated in neurodevelopmental disorders such as autism and intellectual disability, indicating their significant role in both rare and common diseases (Veltman & Brunner, 2012)[8]. Furthermore, genetic variations, including single nucleotide polymorphisms (SNPs), can influence susceptibility to complex diseases like diabetes and heart diseases, as well as impact drug efficacy and adverse drug reactions (Kamatani & Nakamura, 2021)[3].
The high frequency of genetic variations presents a challenge for experimental validation of their functions and interactions. However, advances in computational approaches, particularly machine learning, have significantly improved our ability to predict the functional consequences of mutations, including their effects on protein structure, gene expression, and disease risk (Shea et al., 2023)[1]. These models classify mutations based on their potential impacts, allowing for better understanding and predictions regarding disease mechanisms.
Ethical considerations surrounding genetic testing are increasingly relevant as the implications of identifying genetic mutations become clearer. The potential for genetic testing to reveal predispositions to diseases raises questions about privacy, discrimination, and the psychological impact of knowing one's genetic risks. Moreover, the ethical use of genetic information in clinical settings necessitates careful consideration of informed consent and the potential for genetic discrimination in insurance and employment contexts (Zoghbi & Beaudet, 2016)[48].
In summary, genetic mutations are integral to the understanding of disease mechanisms, influencing both individual health outcomes and broader public health implications. As genetic testing becomes more prevalent, the associated ethical considerations will need to be addressed to ensure responsible use of genetic information in clinical practice and research.
6 Future Directions in Research
6.1 Gene Therapy
Genetic mutations play a pivotal role in the etiology of various diseases, encompassing both inherited and acquired conditions. Germline mutations are primarily responsible for heritable diseases, while somatic mutations are often implicated in cancer and other acquired disorders. Recent research has highlighted the importance of understanding the rates, spectrum, and determinants of these mutations, which can inform our understanding of both Mendelian and complex human diseases [2].
The advancement of genome sequencing technologies has significantly enhanced our ability to identify causal mutations associated with genetic diseases. As a result, model organisms are increasingly utilized to validate specific mutations identified in patients and to explore potential therapeutic targets [37]. The identification of over 3000 genes associated with disease-causing mutations has led to the initiation of approximately 2600 gene therapy trials aimed at managing various disorders [49].
Gene therapy has emerged as a transformative approach to treat genetic diseases by directly addressing the underlying genetic abnormalities. Techniques such as genome editing—utilizing tools like CRISPR/Cas9, Zinc Finger Nucleases (ZFNs), and Transcription Activator-Like Effector Nucleases (TALENs)—allow for targeted manipulation of genomes, offering the potential for curative treatments [50]. This approach not only provides new avenues for treatment but also raises ethical considerations regarding the manipulation of genetic material [51].
In the context of gene therapy, challenges remain, including vector delivery limitations, immunotoxicity, and the need for specificity in targeting the appropriate genetic sequences. Future research directions emphasize the importance of optimizing these therapeutic strategies to ensure long-term effectiveness and safety [51]. Moreover, the potential of microRNAs (miRNAs) as modulators of gene expression presents an additional layer of complexity, as their dysregulation can also contribute to disease phenotypes [52].
The future landscape of gene therapy is poised to evolve further as researchers continue to explore innovative approaches for gene replacement, gene suppression, and gene editing, particularly in the treatment of complex diseases such as cancer and multifactorial conditions [50]. As the field progresses, the integration of genetic insights into clinical practice will be crucial for developing personalized medicine strategies aimed at improving patient outcomes. Thus, the role of genetic mutations in disease not only underscores the importance of continued research in gene therapy but also highlights the potential for these advancements to revolutionize disease management and treatment paradigms.
6.2 Precision Medicine
Genetic mutations play a pivotal role in the understanding and treatment of diseases, particularly within the framework of precision medicine. The core principle of precision medicine is to tailor medical treatment to the individual characteristics of each patient, which is largely informed by genetic variations. These variations can significantly influence disease susceptibility, progression, and response to treatment.
The significance of genetic mutations in disease has been emphasized in several studies. For instance, precision medicine approaches are increasingly being utilized to identify specific genetic variants that underlie various diseases, including cancer and congenital heart disease. The ability to determine these genetic variants has dramatically improved over the past three decades, allowing for the development of tailored therapeutic strategies aimed at enhancing patient care and outcomes [53]. This approach is especially crucial in oncology, where the identification of mutations in genes such as KRAS, BRAF, and PIK3CA can inform the selection of targeted therapies that inhibit specific molecular pathways involved in tumor growth [54].
Moreover, the integration of genetic and epigenetic data is essential for a comprehensive understanding of disease mechanisms. Studies have indicated that the interplay between genetic mutations and epigenetic modifications can influence the development and progression of diseases, such as cancer. This integrated perspective is vital for the advancement of precision medicine, as it allows for a more nuanced understanding of how these factors interact and contribute to disease pathology [55].
The application of next-generation sequencing (NGS) has revolutionized the landscape of precision medicine by enabling the detailed analysis of genetic alterations associated with diseases. This technology facilitates the identification of mutations and the assessment of their implications for treatment response, thereby supporting the paradigm shift from traditional phenotype-based classifications to a more refined nosology based on genetic and biological data [56].
Future research directions in this field are likely to focus on several key areas. Firstly, there is a need for the continued exploration of the functional implications of identified genetic mutations, particularly how they interact with environmental factors and epigenetic changes. Understanding these interactions can enhance the predictive power of genetic testing and improve the customization of therapeutic approaches [57]. Secondly, advancements in artificial intelligence and machine learning can facilitate the integration of multiomics data, leading to more accurate models of disease that can inform treatment decisions [58].
Furthermore, addressing the challenges of genetic diversity in populations and ensuring equitable access to precision medicine will be crucial. As highlighted in discussions about genetic counseling among minority populations, understanding genetic variation and its implications for disease prevention and management is essential for the successful implementation of precision medicine [59].
In summary, genetic mutations are central to the advancement of precision medicine, influencing disease mechanisms and treatment outcomes. Ongoing research will continue to refine our understanding of these mutations, their interactions with other biological factors, and their role in the development of personalized therapeutic strategies.
6.3 Ongoing Clinical Trials
Genetic mutations play a significant role in the development and understanding of various diseases, both rare and common. These mutations can be classified into several categories, including germline variations, somatic mutations, and de novo mutations, each contributing differently to disease pathology and treatment strategies.
Germline variations are inherited genetic changes that can be causative of genetic diseases and are associated with the risk of various disorders. They also influence drug efficacy and the occurrence of adverse drug reactions. For instance, certain germline variants have been linked to the risk of developing specific conditions, thus highlighting their importance in both disease predisposition and pharmacogenomics[3].
Somatic mutations, on the other hand, are acquired changes that occur in non-germline tissues and are often associated with tumorigenesis. These mutations can drive the progression of cancer by altering key signaling pathways and cellular processes, making them critical targets for cancer therapies[3].
De novo mutations, which arise spontaneously rather than being inherited, have been shown to significantly contribute to the burden of genetic diseases, particularly neurodevelopmental disorders such as intellectual disability, autism, and schizophrenia. Recent research utilizing family-based whole-genome and whole-exome sequencing has underscored the prominence of these mutations in the genetic landscape of both rare and common diseases[8].
The integration of genetic mutations into clinical research has paved the way for more personalized medicine approaches. With advancements in genomic technologies, including genome-wide association studies (GWAS) and next-generation sequencing, researchers can better understand how genetic diversity affects disease susceptibility and treatment responses. This knowledge facilitates the identification of molecularly defined targets for drug development and the design of clinical trials that focus on smaller, genetically homogeneous patient populations[47].
Ongoing clinical trials increasingly leverage this genetic insight to classify diseases based on genetic criteria, enabling the development of targeted therapies. However, challenges remain, particularly in the context of rare diseases, where patient populations are small and the natural history of the diseases is often poorly understood. There is a growing need for translational science to address these challenges by improving diagnostic testing, creating patient registries, and innovating trial designs to better accommodate the complexities of genetic variations[60].
In conclusion, genetic mutations play a critical role in the pathogenesis of diseases, influencing both the understanding and treatment of various conditions. The future of research and clinical trials will likely focus on harnessing this genetic information to enhance personalized medicine, improving outcomes for patients across a spectrum of diseases.
7 Conclusion
The exploration of genetic mutations has unveiled critical insights into their role in the etiology of various diseases, from monogenic disorders to complex multifactorial conditions. Key findings indicate that point mutations, insertions and deletions, copy number variations, and chromosomal alterations significantly influence disease mechanisms and progression. The current research landscape emphasizes the necessity of understanding the intricate interplay between genetic and environmental factors, particularly as it pertains to the onset and progression of diseases like cancer, cardiovascular conditions, and neurodevelopmental disorders. Future research directions should focus on harnessing advancements in gene therapy and precision medicine to develop targeted interventions that address the unique genetic underpinnings of individual patients. As we advance our understanding of genetic mutations, the integration of this knowledge into clinical practice will be essential for improving diagnostic accuracy, treatment efficacy, and overall patient outcomes. Furthermore, ethical considerations surrounding genetic testing and its implications for healthcare access and discrimination must be addressed to ensure equitable advancements in the field.
References
- [1] Anthony Shea;Josh Bartz;Lei Zhang;Xiao Dong. Predicting mutational function using machine learning.. Mutation research. Reviews in mutation research(IF=4.2). 2023. PMID:36965820. DOI: 10.1016/j.mrrev.2023.108457.
- [2] Jay Shendure;Joshua M Akey. The origins, determinants, and consequences of human mutations.. Science (New York, N.Y.)(IF=45.8). 2015. PMID:26404824. DOI: 10.1126/science.aaa9119.
- [3] Yoichiro Kamatani;Yusuke Nakamura. Genetic variations in medical research in the past, at present and in the future.. Proceedings of the Japan Academy. Series B, Physical and biological sciences(IF=4.6). 2021. PMID:34121043. DOI: 10.2183/pjab.97.018.
- [4] Anita Joy-Thomas;Zarna Lalwani;Leticia Guajardo;John Valenza;Walid D Fakhouri. The Role of Genetics in Human Oral Health: A Systematic-Narrative Review.. Dentistry journal(IF=3.1). 2025. PMID:40136761. DOI: 10.3390/dj13030133.
- [5] Byungho Lim;Jihyeob Mun;Seon-Young Kim. Intrinsic Molecular Processes: Impact on Mutagenesis.. Trends in cancer(IF=17.5). 2017. PMID:28718413. DOI: 10.1016/j.trecan.2017.03.009.
- [6] Gordon W Duff. Evidence for genetic variation as a factor in maintaining health.. The American journal of clinical nutrition(IF=6.9). 2006. PMID:16470008. DOI: 10.1093/ajcn/83.2.431S.
- [7] Ian Martin;Valina L Dawson;Ted M Dawson. Recent advances in the genetics of Parkinson's disease.. Annual review of genomics and human genetics(IF=7.9). 2011. PMID:21639795. DOI: 10.1146/annurev-genom-082410-101440.
- [8] Joris A Veltman;Han G Brunner. De novo mutations in human genetic disease.. Nature reviews. Genetics(IF=52.0). 2012. PMID:22805709. DOI: 10.1038/nrg3241.
- [9] J Arenas;Y Campos;M A Martín. [Molecular genetics of mitochondrial cytopathologies].. Neurologia (Barcelona, Spain)(IF=3.1). 1995. PMID:8838558. DOI: .
- [10] Katerina Ondraskova;Ravery Sebuyoya;Ludmila Moranova;Jitka Holcakova;Petr Vonka;Roman Hrstka;Martin Bartosik. Electrochemical biosensors for analysis of DNA point mutations in cancer research.. Analytical and bioanalytical chemistry(IF=3.8). 2023. PMID:36289102. DOI: 10.1007/s00216-022-04388-7.
- [11] S K Urhoj;L N Jespersen;M Nissen;L H Mortensen;A-M Nybo Andersen. Advanced paternal age and mortality of offspring under 5 years of age: a register-based cohort study.. Human reproduction (Oxford, England)(IF=6.1). 2014. PMID:24316515. DOI: 10.1093/humrep/det399.
- [12] Luca Cartegni;Jinhua Wang;Zhengwei Zhu;Michael Q Zhang;Adrian R Krainer. ESEfinder: A web resource to identify exonic splicing enhancers.. Nucleic acids research(IF=13.1). 2003. PMID:12824367. DOI: 10.1093/nar/gkg616.
- [13] M Krawczak;J Reiss;D N Cooper. The mutational spectrum of single base-pair substitutions in mRNA splice junctions of human genes: causes and consequences.. Human genetics(IF=3.6). 1992. PMID:1427786. DOI: 10.1007/BF00210743.
- [14] Edwin H Cook;Stephen W Scherer. Copy-number variations associated with neuropsychiatric conditions.. Nature(IF=48.5). 2008. PMID:18923514. DOI: 10.1038/nature07458.
- [15] Rebecca L Pollex;Robert A Hegele. Genomic copy number variation and its potential role in lipoprotein and metabolic phenotypes.. Current opinion in lipidology(IF=4.6). 2007. PMID:17353666. DOI: 10.1097/MOL.0b013e32802e6c12.
- [16] Santhosh Girirajan;Catarina D Campbell;Evan E Eichler. Human copy number variation and complex genetic disease.. Annual review of genetics(IF=8.6). 2011. PMID:21854229. DOI: 10.1146/annurev-genet-102209-163544.
- [17] Steven A McCarroll;David M Altshuler. Copy-number variation and association studies of human disease.. Nature genetics(IF=29.0). 2007. PMID:17597780. DOI: 10.1038/ng2080.
- [18] Kathleen Dixon;Elizabeth Kopras. Genetic alterations and DNA repair in human carcinogenesis.. Seminars in cancer biology(IF=15.7). 2004. PMID:15489137. DOI: 10.1016/j.semcancer.2004.06.007.
- [19] Hooman Ganjavi;David Malkin. Genetics of childhood cancer.. Clinical orthopaedics and related research(IF=4.4). 2002. PMID:12151885. DOI: 10.1097/00003086-200208000-00011.
- [20] Sonja C Stadler;C David Allis. Linking epithelial-to-mesenchymal-transition and epigenetic modifications.. Seminars in cancer biology(IF=15.7). 2012. PMID:22706095. DOI: .
- [21] Lisa Pavinato;Arianna Baggiolini. Oncogenic competence: balancing mutations, cellular state, and microenvironment.. Trends in cancer(IF=17.5). 2025. PMID:39875306. DOI: 10.1016/j.trecan.2025.01.002.
- [22] Thomas Gasser. Molecular pathogenesis of Parkinson disease: insights from genetic studies.. Expert reviews in molecular medicine(IF=5.5). 2009. PMID:19631006. DOI: 10.1017/S1462399409001148.
- [23] Xiuzhi Jia;Li Tan;Sixiu Chen;Ruihan Tang;Wei Chen. Monogenic lupus: Tracing the therapeutic implications from single gene mutations.. Clinical immunology (Orlando, Fla.)(IF=3.8). 2023. PMID:37481012. DOI: 10.1016/j.clim.2023.109699.
- [24] S Rubattu;R Giliberti;M Volpe. Etiology and pathophysiology of stroke as a complex trait.. American journal of hypertension(IF=3.1). 2000. PMID:11041170. DOI: 10.1016/s0895-7061(00)01249-8.
- [25] Yi Zhang;Zhi-Ying Wu. Gene therapy for monogenic disorders: challenges, strategies, and perspectives.. Journal of genetics and genomics = Yi chuan xue bao(IF=7.1). 2024. PMID:37586590. DOI: 10.1016/j.jgg.2023.08.001.
- [26] Laurence Tiret. Gene-environment interaction: a central concept in multifactorial diseases.. The Proceedings of the Nutrition Society(IF=4.5). 2002. PMID:12691175. DOI: 10.1079/pns2002178.
- [27] Marta Bertolaso;Jenny Olsson;Antonio Picardi;Jorge Rakela. Gene therapy and enhancement for diabetes (and other diseases): the multiplicity of considerations.. Diabetes/metabolism research and reviews(IF=6.0). 2010. PMID:20818805. DOI: 10.1002/dmrr.1116.
- [28] Gabriel A Cipolla;Jaqueline C de Oliveira;Amanda Salviano-Silva;Sara C Lobo-Alves;Debora S Lemos;Luana C Oliveira;Tayana S Jucoski;Carolina Mathias;Gabrielle A Pedroso;Erika P Zambalde;Daniela F Gradia. Long Non-Coding RNAs in Multifactorial Diseases: Another Layer of Complexity.. Non-coding RNA(IF=3.0). 2018. PMID:29751665. DOI: 10.3390/ncrna4020013.
- [29] Rachel J Boyd;Dimitri Avramopoulos;Lauren L Jantzie;Andrew S McCallion. Neuroinflammation represents a common theme amongst genetic and environmental risk factors for Alzheimer and Parkinson diseases.. Journal of neuroinflammation(IF=10.1). 2022. PMID:36076238. DOI: 10.1186/s12974-022-02584-x.
- [30] Brennan H Baker;Shaoyi Zhang;Jeremy M Simon;Sarah M McLarnan;Wendy K Chung;Brandon L Pearson. Environmental carcinogens disproportionally mutate genes implicated in neurodevelopmental disorders.. Frontiers in neuroscience(IF=3.2). 2023. PMID:37599994. DOI: 10.3389/fnins.2023.1106573.
- [31] G H Lyman. Risk factors for cancer.. Primary care(IF=4.3). 1992. PMID:1410059. DOI: .
- [32] M D Shelby;J B Bishop;J M Mason;K R Tindall. Fertility, reproduction, and genetic disease: studies on the mutagenic effects of environmental agents on mammalian germ cells.. Environmental health perspectives(IF=9.8). 1993. PMID:8354176. DOI: 10.1289/ehp.93100283.
- [33] Gloria Ravegnini;Gulia Sammarini;Patrizia Hrelia;Sabrina Angelini. Key Genetic and Epigenetic Mechanisms in Chemical Carcinogenesis.. Toxicological sciences : an official journal of the Society of Toxicology(IF=4.1). 2015. PMID:26500287. DOI: 10.1093/toxsci/kfv165.
- [34] W A Suk;G W Collman. Genes and the environment: their impact on children's health.. Environmental health perspectives(IF=9.8). 1998. PMID:9646043. DOI: 10.1289/ehp.98106817.
- [35] F Muggia;T Safra;L Dubeau. BRCA genes: lessons learned from experimental and clinical cancer.. Annals of oncology : official journal of the European Society for Medical Oncology(IF=65.4). 2011. PMID:21285156. DOI: 10.1093/annonc/mdq659.
- [36] Kenneth Walsh;Nalini Raghavachari;Candace Kerr;Alexander G Bick;Steven R Cummings;Todd Druley;Cynthia E Dunbar;Giulio Genovese;Margaret A Goodell;Siddhartha Jaiswal;Jaroslaw Maciejewski;Pradeep Natarajan;Anastasia V Shindyapina;Alan R Shuldiner;Erik B Van Den Akker;Jan Vijg. Clonal Hematopoiesis Analyses in Clinical, Epidemiologic, and Genetic Aging Studies to Unravel Underlying Mechanisms of Age-Related Dysfunction in Humans.. Frontiers in aging(IF=4.3). 2022. PMID:35821803. DOI: 10.3389/fragi.2022.841796.
- [37] Katherine A Strynatka;Michelle C Gurrola-Gal;Jason N Berman;Christopher R McMaster. How Surrogate and Chemical Genetics in Model Organisms Can Suggest Therapies for Human Genetic Diseases.. Genetics(IF=5.1). 2018. PMID:29487144. DOI: 10.1534/genetics.117.300124.
- [38] Min Zhang;Ting Hu;Tianyu Ma;Wei Huang;Yan Wang. Epigenetics and environmental health.. Frontiers of medicine(IF=3.5). 2024. PMID:38806988. DOI: 10.1007/s11684-023-1038-2.
- [39] Chen Xue;Qingfei Chu;Qiuxian Zheng;Shiman Jiang;Zhengyi Bao;Yuanshuai Su;Juan Lu;Lanjuan Li. Role of main RNA modifications in cancer: N6-methyladenosine, 5-methylcytosine, and pseudouridine.. Signal transduction and targeted therapy(IF=52.7). 2022. PMID:35484099. DOI: 10.1038/s41392-022-01003-0.
- [40] Emma Cazaly;Jac Charlesworth;Joanne L Dickinson;Adele F Holloway. Genetic Determinants of Epigenetic Patterns: Providing Insight into Disease.. Molecular medicine (Cambridge, Mass.)(IF=6.4). 2015. PMID:25822796. DOI: 10.2119/molmed.2015.00001.
- [41] Aaina Singh Rathore;Hareram Birla;Saumitra Sen Singh;Walia Zahra;Hagera Dilnashin;Richa Singh;Priyanka Kumari Keshri;Surya Pratap Singh. Epigenetic Modulation in Parkinson's Disease and Potential Treatment Therapies.. Neurochemical research(IF=3.8). 2021. PMID:33900517. DOI: 10.1007/s11064-021-03334-w.
- [42] Nadezhda Camacho-Ordonez;Esteban Ballestar;H Th Marc Timmers;Bodo Grimbacher. What can clinical immunology learn from inborn errors of epigenetic regulators?. The Journal of allergy and clinical immunology(IF=11.2). 2021. PMID:33609625. DOI: 10.1016/j.jaci.2021.01.035.
- [43] Elisa Rumi;Chiara Trotti;Daniele Vanni;Ilaria Carola Casetti;Daniela Pietra;Emanuela Sant'Antonio. The Genetic Basis of Primary Myelofibrosis and Its Clinical Relevance.. International journal of molecular sciences(IF=4.9). 2020. PMID:33255170. DOI: 10.3390/ijms21238885.
- [44] Holger Rumpold;Gerald Webersinke. Molecular pathogenesis of Philadelphia-positive chronic myeloid leukemia - is it all BCR-ABL?. Current cancer drug targets(IF=3.5). 2011. PMID:21062244. DOI: 10.2174/156800911793743619.
- [45] Heidi G Sutherland;Cassie L Albury;Lyn R Griffiths. Advances in genetics of migraine.. The journal of headache and pain(IF=7.9). 2019. PMID:31226929. DOI: 10.1186/s10194-019-1017-9.
- [46] Helen K Brittain;Richard Scott;Ellen Thomas. The rise of the genome and personalised medicine.. Clinical medicine (London, England)(IF=3.9). 2017. PMID:29196356. DOI: 10.7861/clinmedicine.17-6-545.
- [47] Yi-Fan Lu;David B Goldstein;Misha Angrist;Gianpiero Cavalleri. Personalized medicine and human genetic diversity.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2014. PMID:25059740. DOI: .
- [48] Huda Y Zoghbi;Arthur L Beaudet. Epigenetics and Human Disease.. Cold Spring Harbor perspectives in biology(IF=8.4). 2016. PMID:26834142. DOI: .
- [49] Tafere Mulaw Belete. The Current Status of Gene Therapy for the Treatment of Cancer.. Biologics : targets & therapy(IF=3.4). 2021. PMID:33776419. DOI: 10.2147/BTT.S302095.
- [50] Aniruddha Sen;Vijay Singh;Shailendra Dwivedi;Ruchika Agrawal;Akash Bansal;Shashank Shekhar;Sudhir Shyam Kushwaha. Gene Therapy and Gene Editing: Current Trends and Future Prospects of Molecular Medicine.. Current gene therapy(IF=3.3). 2025. PMID:39976035. DOI: 10.2174/0115665232347734250211074241.
- [51] Beibei Qie;Jianghua Tuo;Feilong Chen;Haili Ding;Lei Lyu. Gene therapy for genetic diseases: challenges and future directions.. MedComm(IF=10.7). 2025. PMID:39949979. DOI: 10.1002/mco2.70091.
- [52] Alexandra Henrion-Caude;Muriel Girard;Jeanne Amiel. MicroRNAs in genetic disease: rethinking the dosage.. Current gene therapy(IF=3.3). 2012. PMID:22856604. DOI: 10.2174/156652312802083602.
- [53] Bruce D Gelb. Prospects for precision genetic medicine in congenital heart disease.. Current opinion in genetics & development(IF=3.6). 2022. PMID:36115276. DOI: 10.1016/j.gde.2022.101983.
- [54] Adi Muradi Muhar;Adrian Joshua Velaro;Arya Tjipta Prananda;Sony Eka Nugraha;Princella Halim;Rony Abdi Syahputra. Precision medicine in colorectal cancer: genomics profiling and targeted treatment.. Frontiers in pharmacology(IF=4.8). 2025. PMID:40083375. DOI: 10.3389/fphar.2025.1532971.
- [55] Ryuji Hamamoto;Masaaki Komatsu;Ken Takasawa;Ken Asada;Syuzo Kaneko. Epigenetics Analysis and Integrated Analysis of Multiomics Data, Including Epigenetic Data, Using Artificial Intelligence in the Era of Precision Medicine.. Biomolecules(IF=4.8). 2019. PMID:31905969. DOI: 10.3390/biom10010062.
- [56] Yuan Xue;William R Wilcox. Changing paradigm of cancer therapy: precision medicine by next-generation sequencing.. Cancer biology & medicine(IF=8.4). 2016. PMID:27144059. DOI: 10.28092/j.issn.2095-3941.2016.0003.
- [57] Anjali Tripathi;Vinay Kumar Pandey;Garima Sharma;Ashish Ranjan Sharma;Anam Taufeeq;Abhimanyu Kumar Jha;Jin-Chul Kim. Genomic Insights into Dementia: Precision Medicine and the Impact of Gene-Environment Interaction.. Aging and disease(IF=6.9). 2024. PMID:38607741. DOI: 10.14336/AD.2024.0322.
- [58] Iman Hajirasouliha;Olivier Elemento. Precision medicine and artificial intelligence: overview and relevance to reproductive medicine.. Fertility and sterility(IF=7.0). 2020. PMID:33160512. DOI: 10.1016/j.fertnstert.2020.09.156.
- [59] Chanita H Halbert;Barbara W Harrison. Genetic counseling among minority populations in the era of precision medicine.. American journal of medical genetics. Part C, Seminars in medical genetics(IF=4.4). 2018. PMID:29575517. DOI: 10.1002/ajmg.c.31604.
- [60] Anne R Pariser;William A Gahl. Important role of translational science in rare disease innovation, discovery, and drug development.. Journal of general internal medicine(IF=4.2). 2014. PMID:25029971. DOI: 10.1007/s11606-014-2881-2.
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
Genetic Mutations · Disease Susceptibility · Precision Medicine · Genetic Testing · Environmental Factors
© 2025 MaltSci
