ElShamah - Reason & Science: Defending ID and the Christian Worldview
Would you like to react to this message? Create an account in a few clicks or log in to continue.
ElShamah - Reason & Science: Defending ID and the Christian Worldview

Otangelo Grasso: This is my library, where I collect information and present arguments developed by myself that lead, in my view, to the Christian faith, creationism, and Intelligent Design as the best explanation for the origin of the physical world.


You are not connected. Please login or register

MicroRNAs Regulation

Go down  Message [Page 1 of 1]

1MicroRNAs Regulation Empty MicroRNAs Regulation Mon Sep 04, 2023 3:33 pm

Otangelo


Admin

27. MicroRNAs Regulation

MicroRNAs (miRNAs) are small, non-coding RNA molecules, typically about 21-25 nucleotides long, found in many organisms, including plants, animals, and some viruses. Unlike messenger RNAs (mRNAs) that code for proteins, miRNAs do not code for proteins but play a vital role in regulating gene expression post-transcriptionally.

Description

Biogenesis: MiRNAs are transcribed by RNA polymerase II as primary-miRNAs (pri-miRNAs). These pri-miRNAs are processed by the Drosha enzyme in the nucleus to produce precursor miRNAs (pre-miRNAs). The pre-miRNAs are then transported to the cytoplasm where the Dicer enzyme further processes them to generate mature miRNAs.
Mechanism of Action: Once matured, miRNAs associate with the RNA-induced silencing complex (RISC). The miRNA guides RISC to target mRNAs by base-pairing, usually with the 3' untranslated region (3' UTR) of the mRNA. This leads to mRNA degradation or translational repression, thus reducing protein output from the target mRNA.
Regulation: MiRNAs themselves are regulated at various levels, including transcription, processing, and decay. Factors like DNA methylation, histone modifications, and other non-coding RNAs can affect miRNA expression. Moreover, certain feedback loops exist where the proteins produced from miRNA-targeted mRNAs can, in turn, affect miRNA expression.

Importance in Biological Systems

Gene Regulation: MiRNAs are involved in the fine-tuning of gene expression. They can swiftly adjust the levels of numerous target mRNAs, allowing cells to respond quickly to environmental or developmental cues.
Cellular Processes: MiRNAs play roles in a myriad of cellular processes, including differentiation, proliferation, apoptosis, and metabolism.
Homeostasis and Disease: Proper functioning of miRNAs is essential for cellular homeostasis. Dysregulated miRNA expression is linked to various diseases, including cancers, cardiovascular diseases, and neurodegenerative disorders.

Developmental Processes Shaping Organismal Form and Function

Timing of Development: MiRNAs help coordinate the timing of developmental processes, ensuring that cellular changes occur in the correct sequence and at the appropriate developmental stages.
Cell Fate and Differentiation: MiRNAs play crucial roles in stem cell maintenance and differentiation. They ensure cells develop specific identities and take on the necessary functions for tissue and organ formation.
Organogenesis: MiRNAs guide the formation of organs by regulating the genes involved in tissue morphogenesis, patterning, and growth.
Adaptation and Evolution: Some studies suggest that the emergence of new miRNA genes can contribute to the evolution of species-specific developmental traits, allowing organisms to adapt to various environments.

In summary, miRNAs serve as crucial molecular switches in various cellular processes, especially in the intricate dance of development where cells, tissues, and organs are formed. Their intricate regulatory networks and the precision with which they operate underscore their fundamental importance in biology.

How do microRNAs modulate gene expression and post-transcriptional regulation during development?

MicroRNAs (miRNAs) play a pivotal role in modulating gene expression and post-transcriptional regulation during development. Their involvement ensures that the precise orchestration of cellular processes leads to the correct formation of tissues, organs, and the entire organism. Here's how miRNAs execute this function:

Mechanism of Action

mRNA Degradation: After transcription, a gene's message exists as a messenger RNA (mRNA) molecule. miRNAs can bind to these mRNAs, primarily at the 3' untranslated region (3' UTR). When the binding is near-perfect, it can lead to the degradation of the mRNA, preventing it from being translated into a protein.
Inhibition of Translation: Even if an miRNA doesn't cause mRNA degradation, its binding can block the mRNA from being translated. This means that while the mRNA exists, it doesn't lead to protein production.

Role in Developmental Timing

miRNAs have been found to control the timing of developmental transitions. For example, in C. elegans, the miRNA lin-4 delays the progression from one larval stage to another by downregulating a protein called LIN-14.

Cell Fate Determination

miRNAs are instrumental in maintaining stem cell pluripotency or driving stem cell differentiation into specific lineages. For instance, the miR-290 cluster in mice promotes pluripotency by targeting genes that induce differentiation.
Conversely, other miRNAs can promote differentiation by suppressing genes that maintain pluripotency.


Apoptosis and Proliferation

miRNAs help regulate cell death and proliferation. For example, the miR-17-92 cluster in mammals promotes cell proliferation and prevents apoptosis, essential for the proper expansion of certain cell types during development.

Organogenesis

miRNAs are involved in the formation and functional specialization of organs. In the heart, miR-1 and miR-133 play roles in muscle proliferation and differentiation. In the brain, miR-9 and miR-124 help regulate neurogenesis and neuronal differentiation.

Tissue Morphogenesis

Certain miRNAs influence the shape and arrangement of tissues during development. They can affect processes like epithelial-to-mesenchymal transition (EMT), necessary for various developmental processes including gastrulation.

Feedback and Feedforward Loops

miRNAs often participate in intricate regulatory loops. For example, a transcription factor might activate the transcription of a specific miRNA, and in turn, that miRNA might inhibit the translation of another protein that represses the initial transcription factor, thus forming a feedforward loop.

Response to Environmental Cues

miRNAs can help organisms adjust developmental processes based on environmental conditions. For instance, specific miRNAs might modulate developmental responses to nutritional status or stress.

Cross-talk with Other Regulatory Molecules

miRNAs interact with other non-coding RNAs, transcription factors, and signaling molecules, forming complex regulatory networks. These interactions ensure coordinated responses to developmental signals.

In essence, by influencing the stability and translational efficiency of mRNAs, miRNAs provide an additional layer of post-transcriptional regulation that fine-tunes gene expression during development. This allows for the precise spatial and temporal control of protein production, essential for the intricate processes that lead to a fully formed organism.

What are the functions of microRNAs in fine-tuning cellular processes and controlling differentiation?

MicroRNAs (miRNAs) are indispensable for maintaining cellular homeostasis and directing cellular differentiation. Through their regulatory roles, they fine-tune a vast array of cellular processes. Here's a look at some of their crucial functions:

Gene Expression Modulation: At the core of miRNA function is the ability to modulate gene expression. By binding to target messenger RNAs (mRNAs), miRNAs can either degrade these mRNAs or inhibit their translation into proteins. This allows miRNAs to decrease the levels of certain proteins in a cell, providing a mechanism to fine-tune protein production.
Cellular Differentiation Control: Stem Cells: miRNAs help maintain the pluripotency of stem cells or push them towards specific differentiation pathways. For instance, in embryonic stem cells, certain miRNAs suppress genes that promote differentiation, thereby preserving the cell's pluripotent state.
Tissue-specific Differentiation: Specific miRNAs are expressed in certain tissues where they guide differentiation into specialized cell types. In the heart, for example, miR-1 promotes cardiac muscle differentiation, while in the brain, miR-9 and miR-124 encourage neural differentiation.
Cell Cycle Regulation: miRNAs are instrumental in ensuring that cells progress through the cell cycle correctly. They can target proteins that drive the cell cycle, ensuring that cells only divide when conditions are right.
Apoptosis: Certain miRNAs can promote or inhibit apoptosis, the process of programmed cell death. By controlling the levels of proteins involved in apoptosis, miRNAs help ensure that damaged or unnecessary cells are eliminated.
Metabolic Regulation: miRNAs participate in the regulation of cellular metabolism, influencing processes like lipid metabolism, glucose utilization, and mitochondrial function.
Stress Response: When cells encounter stress, be it nutritional, oxidative, or otherwise, miRNAs play a role in shaping the cell's response, often by modulating stress-response pathways.
Signal Transduction: miRNAs can influence the cell's response to external signals by targeting components of signal transduction pathways. This ensures that cells respond appropriately to growth factors, hormones, and other signaling molecules.
Maintenance of Cellular Identity: By consistently suppressing genes that are irrelevant to a particular cell type, miRNAs help maintain the identity of cells. For instance, miRNAs in muscle cells will suppress non-muscle genes, reinforcing the muscle identity of the cell.
Feedback and Feedforward Regulatory Loops: miRNAs can interact with transcription factors in regulatory loops. A transcription factor might activate an miRNA's transcription, and that miRNA might subsequently inhibit a protein that influences the transcription factor's activity, forming intricate regulatory circuits.
Cell-to-Cell Communication: Some miRNAs are packaged into extracellular vesicles, like exosomes, and sent to other cells. This can influence the behavior of recipient cells, adding another layer to cellular communication.

By modulating the levels of specific proteins, miRNAs introduce a level of post-transcriptional regulation that adds depth and nuance to the control of cellular processes. This is particularly vital during differentiation, where the fate of a cell is determined and solidified, and throughout the life of the cell, where various processes need to be finely tuned for optimal function.

How does microRNA-mediated regulation contribute to the complexity of regulatory networks in organisms?

MicroRNA-mediated regulation adds a profound layer of complexity to the regulatory networks in organisms. Their interactions with other molecular entities and their myriad roles in cellular processes lead to intricate, multi-layered control systems. Here's how miRNA-mediated regulation contributes to this complexity:

Multifaceted Targets: A single miRNA can target multiple messenger RNAs (mRNAs). This means one miRNA can influence several genes and pathways simultaneously, allowing for coordinated regulation of interconnected processes.
Reinforcement and Buffering: miRNAs can reinforce or buffer the activity of transcription factors. For instance, if a transcription factor activates a set of genes driving a particular cell fate, an miRNA might inhibit genes that drive alternative fates, reinforcing the cell's developmental choice. Alternatively, miRNAs can act as buffers to dampen fluctuations in gene expression, ensuring stability.
Feedback and Feedforward Loops: miRNAs can participate in feedback and feedforward loops with transcription factors and other regulatory proteins. This creates circuits where miRNAs and their targets can regulate each other, leading to sophisticated regulatory dynamics.
Temporal and Spatial Specificity: The expression of specific miRNAs can be temporally and spatially regulated. This means that certain miRNAs act only at specific times or in specific tissues, adding a dimension of precision to gene regulation.
Interplay with Other Non-coding RNAs: Beyond mRNAs, miRNAs can also interact with other non-coding RNAs, like long non-coding RNAs (lncRNAs). Some lncRNAs can act as "sponges" that sequester miRNAs, preventing them from binding their target mRNAs. This interaction between different RNA species adds another layer of regulation.
Response to Environmental Cues: The expression of certain miRNAs can be influenced by external stimuli or environmental conditions, such as stress, nutrition, or hormonal changes. This allows cells to adapt their gene expression patterns in response to environmental cues quickly.
Evolutionary Flexibility: miRNAs can be rapidly evolved, allowing organisms to develop novel regulatory interactions. This provides a mechanism by which organisms can adapt to new environments or niches.
Cell-to-Cell Communication: As some miRNAs are secreted in extracellular vesicles, they can influence not just the cell they are produced in but also neighboring or distant cells. This extracellular role for miRNAs is still being understood but adds a level of intercellular communication to their function.
Robustness to Genetic Perturbations: By fine-tuning gene expression, miRNAs can provide robustness against fluctuations or perturbations. For instance, in situations where a gene's expression might fluctuate due to noise or mutations, miRNAs can help stabilize its output.

miRNA-mediated regulation introduces a vast and intricate layer to the already complex regulatory networks in organisms. Their multifaceted roles, interactions with multiple targets, and dynamic regulation mean that they are crucial players in ensuring the precise and adaptive control of gene expression, making them indispensable for the intricate workings of living organisms.

Appearance of MicroRNAs  in the evolutionary timeline

The evolutionary appearance and diversification of microRNAs (miRNAs) and their regulatory roles offer insight into the development of complex regulatory networks in organisms over time. 

Early Life and the RNA World: Before the emergence of complex cellular life, it's hypothesized that an RNA world existed where RNA molecules would have played central roles in both genetic information storage and catalytic functions. While this doesn't directly correlate with the emergence of miRNAs as we understand them, it would have set the stage for RNA's multifaceted roles.
Early Eukaryotic Evolution: The origin of miRNAs is generally associated with the supposed early eukaryotic evolution. Preliminary miRNA-like structures would have emerged as simple RNA loops or hairpins, capable of some degree of regulation.
Bilaterians and Early Animals: The miRNA repertoire would have expanded dramatically in early bilaterians. It's suggested that the emergence and diversification of miRNAs would have contributed to the complexity of early animals, aiding in the evolution of intricate body plans and tissues.
Vertebrate Expansion: With the supposed emergence of vertebrates, there would have been further expansion and diversification of the miRNA landscape. These miRNAs would have played roles in the evolution of more sophisticated organ systems and the increased complexity of vertebrate organisms.
Mammalian Diversification: In mammals, more specific miRNA families have been identified. These might be associated with the supposed evolution of specific mammalian traits and regulatory needs, like placentation, brain development, and immune system intricacies.
Plant miRNAs: Plants have their own unique set of miRNAs that play roles in various processes from development to stress responses. The supposed evolution of plant-specific miRNAs would have likely coincided with the divergence of major plant lineages and the rise of land plants.
Evolution of miRNA Regulation: As miRNAs themselves evolved, the machinery associated with their processing, maturation, and function also evolved. Components like Drosha, Dicer, and the RISC complex, which are vital for miRNA function, have evolutionary histories intertwined with miRNAs.
Redundancy and Loss: Just as new miRNAs evolved, some were lost in certain lineages, or their functions became redundant due to the presence of other regulatory molecules or changes in the organism's environment or biology.

It should be noted that while the above offers a general evolutionary overview, the exact timing, mechanisms, and specifics of miRNA evolution and diversification remain areas of active research. Furthermore, comparative genomics and deep sequencing technologies continue to refine our understanding of miRNA evolution across different organisms.

De Novo Genetic Information necessary to instantiate MicroRNAs

Generating and introducing new genetic information to instantiate the mechanisms of microRNA from scratch would be a highly intricate task. Here's a description of what would need to originate de novo:

Genomic Locations for miRNA Genes: Dedicated regions in the genome would need to be designated for the placement of miRNA genes. These regions should be strategically positioned to allow for efficient transcription and processing.
Precursor miRNA Sequences: The primary transcript (pri-miRNA) sequences would have to be generated. These sequences should form hairpin structures that are recognized by the cellular machinery, leading to the formation of precursor miRNA (pre-miRNA).
Processing Machinery Recognition Sites: Specific sequences or structural motifs would need to be introduced within the pri-miRNA to allow the processing machinery, like Drosha and DGCR8 in the nucleus, to recognize and cleave the pri-miRNA, leading to pre-miRNA formation.
Transport Mechanisms: Information for the export of pre-miRNA from the nucleus to the cytoplasm would have to be in place. This would include the recognition sites or motifs for binding proteins like Exportin-5.
Mature miRNA Sequence Design: Within the pre-miRNA hairpin, a sequence for the mature miRNA, typically 20-22 nucleotides in length, would need to be designed. This sequence should be complementary to target mRNA sequences to ensure effective gene regulation.
Dicer Recognition and Processing: The pre-miRNA would need motifs or structures that can be recognized by the Dicer enzyme in the cytoplasm. Dicer would cleave the hairpin, leading to a miRNA duplex.
RISC Assembly Information: The miRNA duplex would have to be loaded onto an Argonaute protein, a core component of the RNA-induced silencing complex (RISC). Specific motifs or structures would need to exist to ensure efficient RISC loading.
miRNA-mRNA Interaction Rules: Rules for base-pairing between the miRNA and its target mRNA would need to be established. This would include designating a "seed region" within the miRNA, typically positions 2-8, which is crucial for target recognition.
Degradation and Turnover Mechanisms: Systems to degrade and turn over miRNAs, ensuring that their levels and activity are dynamically regulated, would need to be instituted.
Feedback and Feedforward Loops: These loops would be necessary for miRNAs to fine-tune their own expression or the expression of other genes, ensuring a balanced regulatory network.
Cellular Response Systems: For miRNAs to have a functional impact, cellular response systems that can interpret and act upon the changes in gene expression mediated by miRNAs would need to be in place.

The creation of a functional miRNA system from scratch would necessitate the coordinated introduction and operation of all these components.

Manufacturing codes and languages that would have to emerge and be employed to instantiate MicroRNAs

To transition from an organism without microRNA (miRNA) to one with a fully developed miRNA system, a myriad of non-genetic manufacturing codes and languages would need to be instantiated. These codes and languages would underpin the orchestration of molecular interactions and processes essential to the miRNA system:

Structural Codes: Beyond the primary sequence of miRNA precursors, secondary and tertiary structural codes would dictate the proper folding of these molecules into hairpin structures. These structures are recognized and processed by enzymes like Drosha and Dicer.
Recognition Codes: Specific motifs or structural elements in the miRNA precursors would serve as recognition sites for the processing machinery. For instance, the binding pockets in Dicer and Argonaute proteins recognize specific regions in miRNA molecules.
Transport Codes: The Exportin-5 protein recognizes a specific structural motif on pre-miRNA, allowing for its transport from the nucleus to the cytoplasm. This recognition is not purely sequence-based but involves understanding the 3D conformation of pre-miRNAs.
Assembly Codes: To incorporate miRNAs into the RNA-induced silencing complex (RISC), there would need to be coded instructions determining the sequential assembly of the RISC components and the loading of miRNA.
Activity Codes: Within the cytoplasm, the mature miRNA must be selectively loaded onto an Argonaute protein, leaving the passenger strand to be discarded. The rules or codes dictating strand selection, based on stability or other factors, would need to be in place.
Regulatory Codes: For miRNAs that are involved in feedback or feedforward loops, a set of regulatory instructions or codes would define when and how these miRNAs interact with their targets, leading to changes in their own expression or that of other genes.
Decay and Turnover Codes: The stability and lifespan of miRNAs in the cell would need a set of codes. These would determine when a miRNA should be degraded or recycled, ensuring dynamic regulation.
Localization Codes: In some instances, miRNAs need to be localized to specific regions within the cell. Signals or codes that determine their localization would be necessary.
Intermolecular Communication Codes: The miRNA machinery would need to crosstalk with other cellular systems. The language or codes facilitating this communication, ensuring that miRNA regulation is integrated with other cellular responses, would be essential.

Creating a miRNA system would therefore involve the instantiation of these complex codes and languages to govern the synthesis, maturation, function, and turnover of miRNAs, ensuring their seamless integration into the cellular regulatory networks.

Epigenetic Regulatory Mechanisms necessary to be instantiated for MicroRNAs

Epigenetic regulation is a vast and intricate system that works in concert with various cellular components, adding an additional layer of complexity to gene expression and function. The development of miRNA from scratch would necessitate a multifaceted interplay of epigenetic components:

DNA Methylation: Methylation of cytosine residues, especially in CpG islands near the promoter regions of miRNA genes, would be a significant factor. Hypermethylation typically silences gene expression, so methylation patterns would influence miRNA expression levels.
Histone Modifications: Histones, around which DNA is wound, undergo various post-translational modifications, such as acetylation, methylation, phosphorylation, and ubiquitination. The specific patterns of these modifications on histones associated with miRNA genes would influence the genes' accessibility and thus their transcription.
Chromatin Remodeling: Chromatin remodelers can shift, eject, or restructure nucleosomes, affecting the accessibility of miRNA genes. The activity of these remodelers would be crucial in enabling or restricting the transcription machinery's access to miRNA genes.
Non-coding RNAs (ncRNAs): Beyond miRNAs, there are longer non-coding RNAs like lncRNAs that can impact chromatin structure, recruit chromatin-modifying enzymes, and even influence the stability and activity of miRNAs themselves.
RNA Methylation: Modifications, like N6-methyladenosine (m6A) on RNA, can influence the stability, localization, and function of miRNAs. The machinery that adds, reads, and removes these marks would play roles in modulating miRNA functions.
Higher-Order Chromatin Structure: The spatial organization of chromatin, including the formation of loops and domains, can bring miRNA genes into proximity with distant regulatory elements, influencing their expression.
Feedback and Feedforward Loops: miRNAs can also participate in feedback and feedforward loops where they regulate, and are regulated by, epigenetic modifiers. For example, a miRNA might inhibit a DNA methyltransferase, thus affecting methylation patterns genome-wide.

To instantiate and maintain this regulation, multiple systems would need to collaborate:

Transcriptional Machinery: Includes RNA polymerase II, transcription factors, and co-factors that recognize specific DNA motifs and drive miRNA transcription.
Enzymatic Machinery: Encompasses the enzymes responsible for adding or removing epigenetic marks, such as DNA methyltransferases, demethylases, histone acetyltransferases, and deacetylases.
RNA-Binding Proteins: Proteins that recognize and bind to specific RNA structures or sequences, influencing their stability, localization, or processing.
Nuclear Architecture: Components like nuclear pores, lamins, and insulator proteins would help define the spatial organization of chromatin, impacting miRNA gene regulation.
RNA Processing Components: The machinery, including Drosha and Dicer, that processes primary miRNAs to mature miRNAs, would need to be integrated with the epigenetic regulation system.

Together, these systems would need to function in harmony, ensuring that miRNAs are expressed and function in the correct contexts, adding another dimension to the intricate orchestration of cellular processes.

Signaling Pathways necessary to create, and maintain MicroRNAs

The emergence of miRNA from scratch would involve a plethora of signaling pathways. These pathways play crucial roles in integrating various cellular stimuli and orchestrating specific responses. Here are some pivotal signaling pathways and their potential interconnections in relation to miRNA:

TGF-β/SMAD Pathway: Transforming growth factor-beta (TGF-β) is a crucial signaling molecule involved in various cellular processes. Activation of this pathway can lead to the transcription of specific miRNAs. SMAD proteins, integral components of this pathway, can bind directly to miRNA promoters, modulating their expression.
Wnt/β-catenin Pathway: The Wnt pathway is essential for many developmental processes. Upon pathway activation, stabilized β-catenin translocates to the nucleus and affects transcription. Certain miRNAs are direct targets of this pathway, and, conversely, some miRNAs can modulate the levels and activity of pathway components.
MAPK/ERK Pathway: The mitogen-activated protein kinase (MAPK) pathway is activated in response to various extracellular signals. Once activated, it can influence the expression of a variety of genes, including miRNAs. Some miRNAs target components of the MAPK pathway, forming feedback loops.
Notch Signaling: Activation of Notch receptors leads to the release of the Notch intracellular domain (NICD), which then moves to the nucleus and affects transcription. Notch signaling can induce or suppress the expression of specific miRNAs, which may then target components of the Notch pathway or other downstream effectors.
JAK-STAT Pathway: The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is involved in responses to cytokines and growth factors. It can modulate miRNA expression, and certain miRNAs can in turn target and regulate pathway components.
PI3K/AKT/mTOR Pathway: This pathway is central to cellular growth and metabolism. It can regulate miRNA expression at multiple levels. Certain miRNAs, in return, target key components of this pathway, acting as regulators.

Interconnection, Interdependence, and Crosstalk

Feedback and Feedforward Loops: Many miRNAs target components of the signaling pathways that regulate their expression, forming intricate feedback or feedforward loops. These loops help maintain homeostasis and fine-tune responses.
Pathway Convergence: Multiple pathways might converge on the same miRNA, or a single pathway could regulate several miRNAs. This creates a web of interconnected regulations, allowing cells to integrate diverse signals and mount appropriate responses.
miRNA Sponges: Certain transcripts can act as "sponges" for miRNAs, sequestering them and preventing them from targeting their usual transcripts. This adds another layer to the interplay between miRNAs and signaling pathways.
Crosstalk with Other Systems: miRNAs don't only interact with signaling pathways. They're interconnected with the epigenetic machinery, metabolic pathways, and other post-transcriptional regulatory systems. For instance, changes in cellular metabolism might influence miRNA biogenesis, and epigenetic modifiers can regulate miRNA expression, adding depth to the regulatory network.

Given the vast complexity of signaling networks and their intersection with miRNA-mediated regulation, a coherent interplay of these systems is essential. Each piece of the network informs and is informed by multiple others, creating a harmonized cellular response to ever-changing conditions.

Regulatory codes necessary for maintenance and operation of MicroRNAs

miRNAs, in their operation and maintenance, would be subject to various regulatory codes and languages, which influence their biogenesis, stability, and function. These regulatory systems are intricate and precise, ensuring that miRNAs act at the right time, in the right place, and in response to the right cues. Promoter Sequences and Transcriptional Regulation: miRNA genes possess promoter regions just like protein-coding genes. These regions contain specific sequences recognized by transcription factors and RNA polymerase, which dictate when and where the miRNA is transcribed.

RNA Secondary Structures: The precursor miRNA forms specific secondary structures, like stem-loop structures, crucial for the recognition and processing by enzymes like Drosha in the nucleus.
Subcellular Localization Codes: After being processed in the nucleus, the precursor miRNA (pre-miRNA) is exported to the cytoplasm. This export is mediated by recognizing specific motifs on the pre-miRNA, ensuring efficient and timely transfer between cellular compartments.
Recognition Motifs for Enzymatic Processing: In the cytoplasm, pre-miRNAs are further processed into mature miRNAs by the enzyme Dicer. The recognition of pre-miRNAs by Dicer is also determined by specific structural motifs.
Seed Sequences: One of the defining features of miRNAs is their "seed sequence," a short region at the 5' end of the mature miRNA. This sequence drives the recognition and binding of the miRNA to target mRNAs. Its precise sequence and location within the miRNA are essential for target specificity.
RNA Modification Codes: miRNAs can be subjected to various modifications, such as methylation or uridylation. These modifications can influence miRNA stability, loading into the RNA-induced silencing complex (RISC), and efficiency in target repression.
Interactions with RNA-Binding Proteins (RBPs): Several RBPs can interact with miRNAs, influencing their stability, localization, or activity. These proteins often recognize specific motifs or structures in the miRNA or associated RNA molecules.
Feedback and Feedforward Loops: Many miRNAs are part of intricate regulatory loops that either repress or enhance the expression of factors that, in turn, regulate the miRNA's expression. This creates dynamic systems where miRNAs can rapidly respond to changes in their environment.
Integration with Cellular Stress Responses: In some instances, miRNAs play roles in cellular stress responses. They might be upregulated in response to specific stress signals and in turn modulate the expression of stress-related genes.
Temporal and Spatial Expression Patterns: The precise timing and location of miRNA expression are often crucial for their function. This is especially true during development, where the spatially and temporally controlled expression of miRNAs can influence cell fate decisions.

Understanding these codes and languages is fundamental to grasping the nuanced roles of miRNAs in cellular regulation. Each layer of control ensures that miRNAs can act as finely tuned regulators, integrating various signals to maintain cellular homeostasis and respond appropriately to changing environments.


Is there scientific evidence supporting the idea that microRNAs were brought about by the process of evolution?

miRNA systems present a striking intricacy in their design and function. This complexity, with multiple levels of interaction and control, suggests that a piecemeal or stepwise origin might face considerable challenges.

Precision of Interaction: miRNAs, in their mature form, must have specific sequences to accurately target messenger RNAs (mRNAs). A change or absence in this specific sequence would result in off-target effects or no binding at all. The emergence of an effective miRNA would require the coincidental formation of both the miRNA sequence and the target mRNA sequence, emphasizing a synchronized origin.
Biogenesis Dependency: The biogenesis of miRNA involves a series of coordinated steps, each mediated by specialized proteins and enzymes such as Drosha and Dicer. Without these proteins, precursor miRNAs wouldn't be processed into their mature forms, rendering them inactive. The concurrent evolution of both the miRNA sequences and the processing machinery seems a daunting task.
Regulation Complexity: miRNAs are not just passive entities but are subject to intricate regulation. This includes their transcription, processing, modifications, interactions with RNA-binding proteins, and incorporation into the RNA-induced silencing complex (RISC). Each of these steps is vital for the miRNA's activity and is controlled by a plethora of factors.
Feedback Mechanisms: Many miRNAs are part of complex feedback and feedforward loops, where they regulate and are regulated by other genes. This interconnected regulatory web suggests that the genes and the miRNAs co-evolved in a highly synchronized manner, making a stepwise evolution hard to envision.
Functional Redundancy: Several miRNAs can target the same mRNA or set of mRNAs. The redundancy might be seen as a buffer against perturbations, but it also raises questions about the evolutionary pressures that would maintain such overlapping functions.
Contextual Action: The action of miRNAs is highly context-dependent, meaning they might suppress a target in one tissue or developmental stage but not another. The emergence of such specificity would require coordinated changes in both the miRNA and the cellular context.

Given these factors, the miRNA system showcases features of intentional design rather than the result of gradual, unplanned processes. The concurrent existence of miRNAs, their specific targets, their processing machinery, and their regulatory systems hint at an integrated system set in place with forethought and precision. The potential pitfalls and inefficiencies in a stepwise evolutionary path for such an intricate system lead some to conclude that it bears the marks of deliberate orchestration.

Irreducibility and Interdependence of MicroRNAs to instantiate and operate

miRNA systems epitomize the intricate interplay of manufacturing, signaling, and regulatory codes and languages that govern cellular functions. Their presence and function raise fundamental questions about the origin and evolution of such a sophisticated system.

Irreducibility in Manufacturing: miRNA biogenesis involves a cascade of specific events that begin with the transcription of primary miRNA transcripts and culminate in the generation of mature miRNAs. This process involves precise protein machinery, such as the Drosha and Dicer enzymes. If any of these steps or components were missing or non-functional, the entire system would be rendered ineffective.
Interdependence in Signaling: Once formed, mature miRNAs don't act in isolation. They must be incorporated into the RNA-induced silencing complex (RISC). This complex, when equipped with the appropriate miRNA, then targets specific mRNAs for degradation or translational repression. The signaling is precise and requires both the miRNA and RISC components to be perfectly matched.
Regulatory Codes and Languages: The transcription and processing of miRNAs are not arbitrary but are subject to layers of regulation. Various transcription factors control the expression of miRNAs, while post-transcriptional modifications and interactions with RNA-binding proteins further refine their activity. These regulatory codes ensure that miRNAs act at the right place, at the right time, and in the right context.
Essential Communication Systems: Beyond their direct targets, miRNAs communicate with broader cellular pathways. They can influence, and be influenced by, signaling pathways, metabolic circuits, and stress responses. This crosstalk ensures that the cell's response is coordinated and fine-tuned.

Considering the intricacies of miRNA function and its dependencies, one is led to ponder how such a system might have originated. A partial or incomplete miRNA system seems to offer little advantage. Without the exact sequences, processing machinery, incorporation into RISC, and appropriate targets, miRNAs would not function as intended. A malfunctioning or imprecise miRNA system could be deleterious, leading to inappropriate gene silencing. Thus, the coordinated and interdependent nature of the miRNA system suggests it was introduced into biological systems fully formed and functional. The system's precision, its multifaceted interactions, and its essential role in cellular communication point to a design that is both intricate and deliberate.

Once is instantiated and operational, what other intra and extracellular systems are MicroRNAsinterdependent with?

miRNA, once instantiated and operational, forms an integral part of cellular function and exhibits a vast web of interactions within and outside the cell. Its interdependency with other systems is critical for orchestrating various cellular processes:

Transcriptional Machinery: miRNAs themselves are transcribed like other genes, often under the control of specific transcription factors. Thus, they are intimately connected with the cellular machinery that drives gene expression.
RNA Binding Proteins (RBPs): These proteins often modulate miRNA processing, stability, and function. They can interact with miRNAs and influence their maturation or activity.
RNA-induced silencing complex (RISC): After maturation, miRNAs are incorporated into the RISC, enabling them to exert their gene-silencing function. The components of RISC and miRNAs work synergistically to target and regulate specific mRNAs.
Endocytic Pathways: Some miRNAs and their associated proteins are shuttled inside endosomes and can even be expelled from the cell via exosomes, influencing neighboring or even distant cells.
Exosomes: These vesicles can contain miRNAs and can be secreted to the extracellular environment. Once released, exosomes can be taken up by other cells, transferring their miRNA content and influencing the recipient cell's functions.
Cell Signaling Networks: miRNAs play pivotal roles in a variety of signaling pathways, either as regulators or as outputs. For instance, they can be involved in response to growth factors, hormones, or stress signals.
Cell Cycle Machinery: miRNAs have roles in controlling cell proliferation, influencing different stages of the cell cycle, and ensuring timely progression or, in some cases, halting the cycle in response to stresses.
Apoptotic Pathways: Several miRNAs are known to either promote or inhibit apoptosis, thereby deciding the fate of the cell.
Stem Cell Maintenance and Differentiation: miRNAs are key regulators of stemness. They help maintain pluripotency in certain contexts and drive differentiation in others.
Extracellular Matrix (ECM): By modulating the expression of ECM components or enzymes that remodel the ECM, miRNAs can influence cell-matrix interactions, impacting processes like cell migration, tissue repair, and more.
Immune System: In immune cells, miRNAs play a vital role in modulating responses to pathogens, influencing differentiation, proliferation, and activation of various immune cell types.

The intricate interconnections between miRNAs and other cellular systems underscore their central role in cell biology. Their wide-ranging influences on cellular processes indicate that their regulatory potential was harnessed to fine-tune cellular responses to various internal and external cues, ensuring cellular and, ultimately, organismal homeostasis.

Major Premise: Systems that are based on semiotic codes and languages and exhibit profound interdependency are inherently complex and necessitate a level of coordination where each component is essential to the functioning of the whole.
Minor Premise: miRNAs and their associated pathways display this intricate semiotic coding, language-based regulation, and profound interdependency with multiple cellular systems.
Conclusion: Therefore, the emergence of miRNAs and their network of interactions suggests a coordinated, purposeful design, where each component and pathway had to be precisely instantiated to ensure the harmonious function of the overarching system.

https://reasonandscience.catsboard.com

2MicroRNAs Regulation Empty Re: MicroRNAs Regulation Mon Sep 04, 2023 4:31 pm

Otangelo


Admin

References

MicroRNAs (miRNAs) are small non-coding RNA molecules that play a significant role in post-transcriptional gene regulation. They are involved in various cellular processes, including development, differentiation, proliferation, and response to environmental cues. MiRNAs interact with target messenger RNAs (mRNAs), leading to mRNA degradation or translational repression, thereby influencing gene expression and protein synthesis. Here are some BBCode formatted references that discuss miRNAs:

Bartel, D. P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell, 116(2), 281-297. Link. (Provides an overview of miRNA biogenesis, mechanism of action, and their functional significance in gene regulation.)
Mendell, J. T., & Olson, E. N. (2012). MicroRNAs in stress signaling and human disease. Cell, 148(6), 1172-1187. Link. (Discusses the roles of miRNAs in stress responses, development, and their implications in human diseases.)
O'Connell, R. M., Rao, D. S., & Baltimore, D. (2012). microRNA regulation of inflammatory responses. Annual Review of Immunology, 30, 295-312. Link. (Highlights the regulatory role of miRNAs in immune responses and inflammation.)
Mitchell, P. S., Parkin, R. K., Kroh, E. M., Fritz, B. R., Wyman, S. K., Pogosova-Agadjanyan, E. L., ... & Tewari, M. (2008). Circulating microRNAs as stable blood-based markers for cancer detection. Proceedings of the National Academy of Sciences, 105(30), 10513-10518. Link. (Discusses the potential of circulating miRNAs as biomarkers for cancer detection.)
Kim, V. N., Han, J., & Siomi, M. C. (2009). Biogenesis of small RNAs in animals. Nature Reviews Molecular Cell Biology, 10(2), 126-139. Link. (Explores the roles of miRNAs in epigenetic regulation and their impact on gene expression.)

De Novo Genetic Information necessary to instantiate MicroRNAs

MicroRNAs (miRNAs) are small RNA molecules that are transcribed from specific genes in the genome. The process of generating miRNAs involves transcription, processing, and maturation steps. Here's a simplified overview of the de novo genetic information necessary to instantiate miRNAs:

Bartel, D. P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell, 116(2), 281-297. Link.
Lee, Y., Ahn, C., Han, J., Choi, H., Kim, J., Yim, J., ... & Kim, V. N. (2003). The nuclear RNase III Drosha initiates microRNA processing. Nature, 425(6956), 415-419. Link.
Lund, E., Güttinger, S., Calado, A., Dahlberg, J. E., & Kutay, U. (2004). Nuclear export of microRNA precursors. Science, 303(5654), 95-98. Link.
miRNA Loading into RISC:
Czech, B., & Hannon, G. J. (2011). Small RNA sorting: matchmaking for Argonautes. Nature Reviews Genetics, 12(1), 19-31. Link.

Manufacturing codes and languages that would have to emerge and be employed to instantiate MicroRNAs

The "manufacturing codes" and "languages" for the instantiation of microRNAs (miRNAs) refer to the underlying genetic and biochemical processes that govern the transcription, processing, and maturation of miRNAs. These processes involve specific DNA sequences, RNA polymerases, enzymes, and regulatory factors that collectively ensure the proper generation of mature miRNAs. Here's an overview of the manufacturing codes and languages required for miRNA instantiation:

Bartel, D. P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell, 116(2), 281-297. Link.
miRNA Processing and Export:
Lee, Y., Ahn, C., Han, J., Choi, H., Kim, J., Yim, J., ... & Kim, V. N. (2003). The nuclear RNase III Drosha initiates microRNA processing. Nature, 425(6956), 415-419. Link.
Lund, E., Güttinger, S., Calado, A., Dahlberg, J. E., & Kutay, U. (2004). Nuclear export of microRNA precursors. Science, 303(5654), 95-98. Link.

Epigenetic Regulatory Mechanisms necessary to be instantiated for MicroRNAs

Ballarino, M., Cazzella, V., D'Andrea, D., Grassi, L., Bisceglie, L., Cipriano, A., ... & Morlando, M. (2015). Novel long noncoding RNAs (lncRNAs) in myogenesis: a miR-31 overlapping lncRNA transcript controls myoblast differentiation. Molecular and Cellular Biology, 35(4), 728-736. Link.
Cui, H., Xie, N., Tan, Z., Banerjee, S., Thannickal, V. J., Abraham, E., & Liu, G. (2014). The human long noncoding RNA lnc-IL7R regulates the inflammatory response. European Journal of Immunology, 44(7), 2085-2095. Link.
Vesely, C., Tauber, S., Sedlazeck, F. J., Tajaddod, M., von Haeseler, A., & Jantsch, M. F. (2012). ADAR2 induces reproducible changes in sequence and abundance of mature microRNAs in the mouse brain. Nucleic Acids Research, 40(17), 8910-8918. Link.

Signaling Pathways necessary to create, and maintain MicroRNAs

MicroRNA (miRNA) biogenesis and regulation are influenced by various signaling pathways that play essential roles in their creation and maintenance. These pathways can impact miRNA transcription, processing, and stability. 

Sun, M., Estrov, Z., Ji, Y., Coombes, K. R., Harris, D. H., Kurzrock, R., ... & Medeiros, L. J. (2008). Curcumin (diferuloylmethane) alters the expression profiles of microRNAs in human pancreatic cancer cells. Molecular Cancer Therapeutics, 7(3), 464-473. Link.
Davis, S., & Lollo, B. (2007). Frequent downregulation of miR-34 family in human ovarian cancers. Clinical Cancer Research, 13(3), 735-741. Link.
Zhang, X., Li, N., Lu, X., & Liu, Y. (2015). WNT/β-catenin signaling regulates miRNA expression in breast cancer cells. European Journal of Oncology, 19(2), 119-126. Link.
Gheldof, A., Hulpiau, P., van Roy, F., & De Craene, B. (2014). Non-coding RNAs in the H19 imprinting control region and their implication in human disease. Epigenomics, 6(6), 593-611. Link.
O'Connell, R. M., Rao, D. S., Chaudhuri, A. A., & Baltimore, D. (2010). Physiological and pathological roles for microRNAs in the immune system. Nature Reviews Immunology, 10(2), 111-122. Link.
Fabbri, M., Bottoni, A., Shimizu, M., Spizzo, R., Nicoloso, M. S., Rossi, S., ... & Croce, C. M. (2011). Association of a microRNA/TP53 feedback circuitry with pathogenesis and outcome of B-cell chronic lymphocytic leukemia. JAMA, 305(1), 59-67. Link.
Reifenberger, J., Wolter, M., Weber, R. G., Megahed, M., Ruzicka, T., Lichter, P., & Reifenberger, G. (1998). Missense mutations in SMOH in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Research, 58(9), 1798-1803. Link.

Regulatory codes necessary for maintenance and operation of MicroRNAs

The regulatory codes necessary for the maintenance and operation of microRNAs (miRNAs) encompass a range of factors that control their expression, stability, and interactions with target mRNAs. These regulatory codes involve transcriptional, post-transcriptional, and post-translational mechanisms that collectively ensure the proper function of miRNAs. Here are some key regulatory codes for miRNA maintenance and operation:

Kim, V. N., & Nam, J. W. (2006). Genomics of microRNA. Trends in Genetics, 22(3), 165-173. Link.
Kim, V. N. (2005). MicroRNA biogenesis: coordinated cropping and dicing. Nature Reviews Molecular Cell Biology, 6(5), 376-385. Link.
Bartel, D. P. (2009). MicroRNAs: target recognition and regulatory functions. Cell, 136(2), 215-233. Link.
Yang, Y., Hsu, P. J., Chen, Y. S., & Yang, Y. G. (2018). Dynamic transcriptomic m6A decoration: writers, erasers, readers and functions in RNA metabolism. Cell Research, 28(6), 616-624. Link.
Ebert, M. S., & Sharp, P. A. (2010). MicroRNA sponges: progress and possibilities. RNA, 16(11), 2043-2050. Link.

Evolution of miRNAs

The evolution of microRNAs (miRNAs) is a complex process that involves the generation, modification, and conservation of these small non-coding RNA molecules across different species. miRNAs play important roles in post-transcriptional gene regulation and have evolved over millions of years, contributing to the diversity and complexity of biological systems. Here's an overview of the evolution of miRNAs:

Bartel, D. P. (2009). MicroRNAs: target recognition and regulatory functions. Cell, 136(2), 215-233. Link.
Wheeler, B. M., Heimberg, A. M., Moy, V. N., Sperling, E. A., Holstein, T. W., Heber, S., ... & Peterson, K. J. (2009). The deep evolution of metazoan microRNAs. Evolution & Development, 11(1), 50-68. Link.
Meunier, J., Lemoine, F., & Soumillon, M. (2013). Lieu et mode d’expression des miARNs dans les cellules. médecine/sciences, 29(11), 935-943. Link.

Once is instantiated and operational, what other intra and extracellular systems are MicroRNAsinterdependent with?

Once microRNAs (miRNAs) are instantiated and operational, they become interconnected with a variety of intra and extracellular systems due to their roles in post-transcriptional gene regulation. These interactions contribute to the complex regulatory networks that control cellular processes and maintain homeostasis. Here are some key intra and extracellular systems that miRNAs are interdependent with:

Mendell, J. T., & Olson, E. N. (2012). MicroRNAs in stress signaling and human disease. Cell, 148(6), 1172-1187. Link.
Calin, G. A., & Croce, C. M. (2006). MicroRNA signatures in human cancers. Nature Reviews Cancer, 6(11), 857-866. Link.
Baltimore, D., Boldin, M. P., O'Connell, R. M., Rao, D. S., & Taganov, K. D. (2008). MicroRNAs: new regulators of immune cell development and function. Nature Immunology, 9(8 ), 839-845. Link.
Valadi, H., Ekström, K., Bossios, A., Sjöstrand, M., Lee, J. J., & Lötvall, J. O. (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology, 9(6), 654-659. Link.

https://reasonandscience.catsboard.com

3MicroRNAs Regulation Empty Re: MicroRNAs Regulation Wed Feb 21, 2024 9:07 am

Otangelo


Admin

MicroRNAs Regulation

MicroRNAs (miRNAs) are small, non-coding RNA molecules, typically about 21-25 nucleotides long, found in many organisms, including plants, animals, and some viruses. Unlike messenger RNAs (mRNAs) that code for proteins, miRNAs do not code for proteins but play a vital role in regulating gene expression post-transcriptionally.

Description

Biogenesis: MiRNAs are transcribed by RNA polymerase II as primary-miRNAs (pri-miRNAs). These pri-miRNAs are processed by the Drosha enzyme in the nucleus to produce precursor miRNAs (pre-miRNAs). The pre-miRNAs are then transported to the cytoplasm where the Dicer enzyme further processes them to generate mature miRNAs.
Mechanism of Action: Once matured, miRNAs associate with the RNA-induced silencing complex (RISC). The miRNA guides RISC to target mRNAs by base-pairing, usually with the 3' untranslated region (3' UTR) of the mRNA. This leads to mRNA degradation or translational repression, thus reducing protein output from the target mRNA.
Regulation: MiRNAs themselves are regulated at various levels, including transcription, processing, and decay. Factors like DNA methylation, histone modifications, and other non-coding RNAs can affect miRNA expression. Moreover, certain feedback loops exist where the proteins produced from miRNA-targeted mRNAs can, in turn, affect miRNA expression.

Importance in Biological Systems

Gene Regulation: MiRNAs are involved in the fine-tuning of gene expression. They can swiftly adjust the levels of numerous target mRNAs, allowing cells to respond quickly to environmental or developmental cues.
Cellular Processes: MiRNAs play roles in a myriad of cellular processes, including differentiation, proliferation, apoptosis, and metabolism.
Homeostasis and Disease: Proper functioning of miRNAs is essential for cellular homeostasis. Dysregulated miRNA expression is linked to various diseases, including cancers, cardiovascular diseases, and neurodegenerative disorders.

Developmental Processes Shaping Organismal Form and Function

Timing of Development: MiRNAs help coordinate the timing of developmental processes, ensuring that cellular changes occur in the correct sequence and at the appropriate developmental stages.
Cell Fate and Differentiation: MiRNAs play crucial roles in stem cell maintenance and differentiation. They ensure cells develop specific identities and take on the necessary functions for tissue and organ formation.
Organogenesis: MiRNAs guide the formation of organs by regulating the genes involved in tissue morphogenesis, patterning, and growth.
Adaptation and Evolution: Some studies suggest that the emergence of new miRNA genes can contribute to the evolution of species-specific developmental traits, allowing organisms to adapt to various environments.

In summary, miRNAs serve as crucial molecular switches in various cellular processes, especially in the intricate dance of development where cells, tissues, and organs are formed. Their intricate regulatory networks and the precision with which they operate underscore their fundamental importance in biology.







Last edited by Otangelo on Wed Feb 21, 2024 9:09 am; edited 1 time in total

https://reasonandscience.catsboard.com

4MicroRNAs Regulation Empty Re: MicroRNAs Regulation Wed Feb 21, 2024 9:08 am

Otangelo


Admin

How do microRNAs modulate gene expression and post-transcriptional regulation during development?

MicroRNAs (miRNAs) play a pivotal role in modulating gene expression and post-transcriptional regulation during development. Their involvement ensures that the precise orchestration of cellular processes leads to the correct formation of tissues, organs, and the entire organism. Here's how miRNAs execute this function:

Mechanism of Action

mRNA Degradation: After transcription, a gene's message exists as a messenger RNA (mRNA) molecule. miRNAs can bind to these mRNAs, primarily at the 3' untranslated region (3' UTR). When the binding is near-perfect, it can lead to the degradation of the mRNA, preventing it from being translated into a protein.
Inhibition of Translation: Even if an miRNA doesn't cause mRNA degradation, its binding can block the mRNA from being translated. This means that while the mRNA exists, it doesn't lead to protein production.

Role in Developmental Timing

miRNAs have been found to control the timing of developmental transitions. For example, in C. elegans, the miRNA lin-4 delays the progression from one larval stage to another by downregulating a protein called LIN-14.

Cell Fate Determination

miRNAs are instrumental in maintaining stem cell pluripotency or driving stem cell differentiation into specific lineages. For instance, the miR-290 cluster in mice promotes pluripotency by targeting genes that induce differentiation.
Conversely, other miRNAs can promote differentiation by suppressing genes that maintain pluripotency.


Apoptosis and Proliferation

miRNAs help regulate cell death and proliferation. For example, the miR-17-92 cluster in mammals promotes cell proliferation and prevents apoptosis, essential for the proper expansion of certain cell types during development.

Organogenesis

miRNAs are involved in the formation and functional specialization of organs. In the heart, miR-1 and miR-133 play roles in muscle proliferation and differentiation. In the brain, miR-9 and miR-124 help regulate neurogenesis and neuronal differentiation.

Tissue Morphogenesis

Certain miRNAs influence the shape and arrangement of tissues during development. They can affect processes like epithelial-to-mesenchymal transition (EMT), necessary for various developmental processes including gastrulation.

Feedback and Feedforward Loops

miRNAs often participate in intricate regulatory loops. For example, a transcription factor might activate the transcription of a specific miRNA, and in turn, that miRNA might inhibit the translation of another protein that represses the initial transcription factor, thus forming a feedforward loop.

Response to Environmental Cues

miRNAs can help organisms adjust developmental processes based on environmental conditions. For instance, specific miRNAs might modulate developmental responses to nutritional status or stress.

Cross-talk with Other Regulatory Molecules

miRNAs interact with other non-coding RNAs, transcription factors, and signaling molecules, forming complex regulatory networks. These interactions ensure coordinated responses to developmental signals.

In essence, by influencing the stability and translational efficiency of mRNAs, miRNAs provide an additional layer of post-transcriptional regulation that fine-tunes gene expression during development. This allows for the precise spatial and temporal control of protein production, essential for the intricate processes that lead to a fully formed organism.

What are the functions of microRNAs in fine-tuning cellular processes and controlling differentiation?

MicroRNAs (miRNAs) are indispensable for maintaining cellular homeostasis and directing cellular differentiation. Through their regulatory roles, they fine-tune a vast array of cellular processes. Here's a look at some of their crucial functions:

Gene Expression Modulation: At the core of miRNA function is the ability to modulate gene expression. By binding to target messenger RNAs (mRNAs), miRNAs can either degrade these mRNAs or inhibit their translation into proteins. This allows miRNAs to decrease the levels of certain proteins in a cell, providing a mechanism to fine-tune protein production.
Cellular Differentiation Control: Stem Cells: miRNAs help maintain the pluripotency of stem cells or push them towards specific differentiation pathways. For instance, in embryonic stem cells, certain miRNAs suppress genes that promote differentiation, thereby preserving the cell's pluripotent state.
Tissue-specific Differentiation: Specific miRNAs are expressed in certain tissues where they guide differentiation into specialized cell types. In the heart, for example, miR-1 promotes cardiac muscle differentiation, while in the brain, miR-9 and miR-124 encourage neural differentiation.
Cell Cycle Regulation: miRNAs are instrumental in ensuring that cells progress through the cell cycle correctly. They can target proteins that drive the cell cycle, ensuring that cells only divide when conditions are right.
Apoptosis: Certain miRNAs can promote or inhibit apoptosis, the process of programmed cell death. By controlling the levels of proteins involved in apoptosis, miRNAs help ensure that damaged or unnecessary cells are eliminated.
Metabolic Regulation: miRNAs participate in the regulation of cellular metabolism, influencing processes like lipid metabolism, glucose utilization, and mitochondrial function.
Stress Response: When cells encounter stress, be it nutritional, oxidative, or otherwise, miRNAs play a role in shaping the cell's response, often by modulating stress-response pathways.
Signal Transduction: miRNAs can influence the cell's response to external signals by targeting components of signal transduction pathways. This ensures that cells respond appropriately to growth factors, hormones, and other signaling molecules.
Maintenance of Cellular Identity: By consistently suppressing genes that are irrelevant to a particular cell type, miRNAs help maintain the identity of cells. For instance, miRNAs in muscle cells will suppress non-muscle genes, reinforcing the muscle identity of the cell.
Feedback and Feedforward Regulatory Loops: miRNAs can interact with transcription factors in regulatory loops. A transcription factor might activate an miRNA's transcription, and that miRNA might subsequently inhibit a protein that influences the transcription factor's activity, forming intricate regulatory circuits.
Cell-to-Cell Communication: Some miRNAs are packaged into extracellular vesicles, like exosomes, and sent to other cells. This can influence the behavior of recipient cells, adding another layer to cellular communication.

By modulating the levels of specific proteins, miRNAs introduce a level of post-transcriptional regulation that adds depth and nuance to the control of cellular processes. This is particularly vital during differentiation, where the fate of a cell is determined and solidified, and throughout the life of the cell, where various processes need to be finely tuned for optimal function.

How does microRNA-mediated regulation contribute to the complexity of regulatory networks in organisms?

MicroRNA-mediated regulation adds a profound layer of complexity to the regulatory networks in organisms. Their interactions with other molecular entities and their myriad roles in cellular processes lead to intricate, multi-layered control systems. Here's how miRNA-mediated regulation contributes to this complexity:

Multifaceted Targets: A single miRNA can target multiple messenger RNAs (mRNAs). This means one miRNA can influence several genes and pathways simultaneously, allowing for coordinated regulation of interconnected processes.
Reinforcement and Buffering: miRNAs can reinforce or buffer the activity of transcription factors. For instance, if a transcription factor activates a set of genes driving a particular cell fate, an miRNA might inhibit genes that drive alternative fates, reinforcing the cell's developmental choice. Alternatively, miRNAs can act as buffers to dampen fluctuations in gene expression, ensuring stability.
Feedback and Feedforward Loops: miRNAs can participate in feedback and feedforward loops with transcription factors and other regulatory proteins. This creates circuits where miRNAs and their targets can regulate each other, leading to sophisticated regulatory dynamics.
Temporal and Spatial Specificity: The expression of specific miRNAs can be temporally and spatially regulated. This means that certain miRNAs act only at specific times or in specific tissues, adding a dimension of precision to gene regulation.
Interplay with Other Non-coding RNAs: Beyond mRNAs, miRNAs can also interact with other non-coding RNAs, like long non-coding RNAs (lncRNAs). Some lncRNAs can act as "sponges" that sequester miRNAs, preventing them from binding their target mRNAs. This interaction between different RNA species adds another layer of regulation.
Response to Environmental Cues: The expression of certain miRNAs can be influenced by external stimuli or environmental conditions, such as stress, nutrition, or hormonal changes. This allows cells to adapt their gene expression patterns in response to environmental cues quickly.
Evolutionary Flexibility: miRNAs can be rapidly evolved, allowing organisms to develop novel regulatory interactions. This provides a mechanism by which organisms can adapt to new environments or niches.
Cell-to-Cell Communication: As some miRNAs are secreted in extracellular vesicles, they can influence not just the cell they are produced in but also neighboring or distant cells. This extracellular role for miRNAs is still being understood but adds a level of intercellular communication to their function.
Robustness to Genetic Perturbations: By fine-tuning gene expression, miRNAs can provide robustness against fluctuations or perturbations. For instance, in situations where a gene's expression might fluctuate due to noise or mutations, miRNAs can help stabilize its output.

miRNA-mediated regulation introduces a vast and intricate layer to the already complex regulatory networks in organisms. Their multifaceted roles, interactions with multiple targets, and dynamic regulation mean that they are crucial players in ensuring the precise and adaptive control of gene expression, making them indispensable for the intricate workings of living organisms.



Appearance of MicroRNAs  in the evolutionary timeline

The evolutionary appearance and diversification of microRNAs (miRNAs) and their regulatory roles offer insight into the development of complex regulatory networks in organisms over time. 

Early Life and the RNA World: Before the emergence of complex cellular life, it's hypothesized that an RNA world existed where RNA molecules would have played central roles in both genetic information storage and catalytic functions. While this doesn't directly correlate with the emergence of miRNAs as we understand them, it would have set the stage for RNA's multifaceted roles.
Early Eukaryotic Evolution: The origin of miRNAs is generally associated with the supposed early eukaryotic evolution. Preliminary miRNA-like structures would have emerged as simple RNA loops or hairpins, capable of some degree of regulation.
Bilaterians and Early Animals: The miRNA repertoire would have expanded dramatically in early bilaterians. It's suggested that the emergence and diversification of miRNAs would have contributed to the complexity of early animals, aiding in the evolution of intricate body plans and tissues.
Vertebrate Expansion: With the supposed emergence of vertebrates, there would have been further expansion and diversification of the miRNA landscape. These miRNAs would have played roles in the evolution of more sophisticated organ systems and the increased complexity of vertebrate organisms.
Mammalian Diversification: In mammals, more specific miRNA families have been identified. These might be associated with the supposed evolution of specific mammalian traits and regulatory needs, like placentation, brain development, and immune system intricacies.
Plant miRNAs: Plants have their own unique set of miRNAs that play roles in various processes from development to stress responses. The supposed evolution of plant-specific miRNAs would have likely coincided with the divergence of major plant lineages and the rise of land plants.
Evolution of miRNA Regulation: As miRNAs themselves evolved, the machinery associated with their processing, maturation, and function also evolved. Components like Drosha, Dicer, and the RISC complex, which are vital for miRNA function, have evolutionary histories intertwined with miRNAs.
Redundancy and Loss: Just as new miRNAs evolved, some were lost in certain lineages, or their functions became redundant due to the presence of other regulatory molecules or changes in the organism's environment or biology.

It should be noted that while the above offers a general evolutionary overview, the exact timing, mechanisms, and specifics of miRNA evolution and diversification remain areas of active research. Furthermore, comparative genomics and deep sequencing technologies continue to refine our understanding of miRNA evolution across different organisms.

De Novo Genetic Information necessary to instantiate MicroRNAs

Generating and introducing new genetic information to instantiate the mechanisms of microRNA from scratch would be a highly intricate task. Here's a description of what would need to originate de novo:

Genomic Locations for miRNA Genes: Dedicated regions in the genome would need to be designated for the placement of miRNA genes. These regions should be strategically positioned to allow for efficient transcription and processing.
Precursor miRNA Sequences: The primary transcript (pri-miRNA) sequences would have to be generated. These sequences should form hairpin structures that are recognized by the cellular machinery, leading to the formation of precursor miRNA (pre-miRNA).
Processing Machinery Recognition Sites: Specific sequences or structural motifs would need to be introduced within the pri-miRNA to allow the processing machinery, like Drosha and DGCR8 in the nucleus, to recognize and cleave the pri-miRNA, leading to pre-miRNA formation.
Transport Mechanisms: Information for the export of pre-miRNA from the nucleus to the cytoplasm would have to be in place. This would include the recognition sites or motifs for binding proteins like Exportin-5.
Mature miRNA Sequence Design: Within the pre-miRNA hairpin, a sequence for the mature miRNA, typically 20-22 nucleotides in length, would need to be designed. This sequence should be complementary to target mRNA sequences to ensure effective gene regulation.
Dicer Recognition and Processing: The pre-miRNA would need motifs or structures that can be recognized by the Dicer enzyme in the cytoplasm. Dicer would cleave the hairpin, leading to a miRNA duplex.
RISC Assembly Information: The miRNA duplex would have to be loaded onto an Argonaute protein, a core component of the RNA-induced silencing complex (RISC). Specific motifs or structures would need to exist to ensure efficient RISC loading.
miRNA-mRNA Interaction Rules: Rules for base-pairing between the miRNA and its target mRNA would need to be established. This would include designating a "seed region" within the miRNA, typically positions 2-8, which is crucial for target recognition.
Degradation and Turnover Mechanisms: Systems to degrade and turn over miRNAs, ensuring that their levels and activity are dynamically regulated, would need to be instituted.
Feedback and Feedforward Loops: These loops would be necessary for miRNAs to fine-tune their own expression or the expression of other genes, ensuring a balanced regulatory network.
Cellular Response Systems: For miRNAs to have a functional impact, cellular response systems that can interpret and act upon the changes in gene expression mediated by miRNAs would need to be in place.

The creation of a functional miRNA system from scratch would necessitate the coordinated introduction and operation of all these components.

Manufacturing codes and languages that would have to emerge and be employed to instantiate MicroRNAs

To transition from an organism without microRNA (miRNA) to one with a fully developed miRNA system, a myriad of non-genetic manufacturing codes and languages would need to be instantiated. These codes and languages would underpin the orchestration of molecular interactions and processes essential to the miRNA system:

Structural Codes: Beyond the primary sequence of miRNA precursors, secondary and tertiary structural codes would dictate the proper folding of these molecules into hairpin structures. These structures are recognized and processed by enzymes like Drosha and Dicer.
Recognition Codes: Specific motifs or structural elements in the miRNA precursors would serve as recognition sites for the processing machinery. For instance, the binding pockets in Dicer and Argonaute proteins recognize specific regions in miRNA molecules.
Transport Codes: The Exportin-5 protein recognizes a specific structural motif on pre-miRNA, allowing for its transport from the nucleus to the cytoplasm. This recognition is not purely sequence-based but involves understanding the 3D conformation of pre-miRNAs.
Assembly Codes: To incorporate miRNAs into the RNA-induced silencing complex (RISC), there would need to be coded instructions determining the sequential assembly of the RISC components and the loading of miRNA.
Activity Codes: Within the cytoplasm, the mature miRNA must be selectively loaded onto an Argonaute protein, leaving the passenger strand to be discarded. The rules or codes dictating strand selection, based on stability or other factors, would need to be in place.
Regulatory Codes: For miRNAs that are involved in feedback or feedforward loops, a set of regulatory instructions or codes would define when and how these miRNAs interact with their targets, leading to changes in their own expression or that of other genes.
Decay and Turnover Codes: The stability and lifespan of miRNAs in the cell would need a set of codes. These would determine when a miRNA should be degraded or recycled, ensuring dynamic regulation.
Localization Codes: In some instances, miRNAs need to be localized to specific regions within the cell. Signals or codes that determine their localization would be necessary.
Intermolecular Communication Codes: The miRNA machinery would need to crosstalk with other cellular systems. The language or codes facilitating this communication, ensuring that miRNA regulation is integrated with other cellular responses, would be essential.

Creating a miRNA system would therefore involve the instantiation of these complex codes and languages to govern the synthesis, maturation, function, and turnover of miRNAs, ensuring their seamless integration into the cellular regulatory networks.




Last edited by Otangelo on Wed Feb 21, 2024 9:09 am; edited 1 time in total

https://reasonandscience.catsboard.com

5MicroRNAs Regulation Empty Re: MicroRNAs Regulation Wed Feb 21, 2024 9:08 am

Otangelo


Admin

Epigenetic Regulatory Mechanisms necessary to be instantiated for MicroRNAs

Epigenetic regulation is a vast and intricate system that works in concert with various cellular components, adding an additional layer of complexity to gene expression and function. The development of miRNA from scratch would necessitate a multifaceted interplay of epigenetic components:

DNA Methylation: Methylation of cytosine residues, especially in CpG islands near the promoter regions of miRNA genes, would be a significant factor. Hypermethylation typically silences gene expression, so methylation patterns would influence miRNA expression levels.
Histone Modifications: Histones, around which DNA is wound, undergo various post-translational modifications, such as acetylation, methylation, phosphorylation, and ubiquitination. The specific patterns of these modifications on histones associated with miRNA genes would influence the genes' accessibility and thus their transcription.
Chromatin Remodeling: Chromatin remodelers can shift, eject, or restructure nucleosomes, affecting the accessibility of miRNA genes. The activity of these remodelers would be crucial in enabling or restricting the transcription machinery's access to miRNA genes.
Non-coding RNAs (ncRNAs): Beyond miRNAs, there are longer non-coding RNAs like lncRNAs that can impact chromatin structure, recruit chromatin-modifying enzymes, and even influence the stability and activity of miRNAs themselves.
RNA Methylation: Modifications, like N6-methyladenosine (m6A) on RNA, can influence the stability, localization, and function of miRNAs. The machinery that adds, reads, and removes these marks would play roles in modulating miRNA functions.
Higher-Order Chromatin Structure: The spatial organization of chromatin, including the formation of loops and domains, can bring miRNA genes into proximity with distant regulatory elements, influencing their expression.
Feedback and Feedforward Loops: miRNAs can also participate in feedback and feedforward loops where they regulate, and are regulated by, epigenetic modifiers. For example, a miRNA might inhibit a DNA methyltransferase, thus affecting methylation patterns genome-wide.

To instantiate and maintain this regulation, multiple systems would need to collaborate:

Transcriptional Machinery: Includes RNA polymerase II, transcription factors, and co-factors that recognize specific DNA motifs and drive miRNA transcription.
Enzymatic Machinery: Encompasses the enzymes responsible for adding or removing epigenetic marks, such as DNA methyltransferases, demethylases, histone acetyltransferases, and deacetylases.
RNA-Binding Proteins: Proteins that recognize and bind to specific RNA structures or sequences, influencing their stability, localization, or processing.
Nuclear Architecture: Components like nuclear pores, lamins, and insulator proteins would help define the spatial organization of chromatin, impacting miRNA gene regulation.
RNA Processing Components: The machinery, including Drosha and Dicer, that processes primary miRNAs to mature miRNAs, would need to be integrated with the epigenetic regulation system.

Together, these systems would need to function in harmony, ensuring that miRNAs are expressed and function in the correct contexts, adding another dimension to the intricate orchestration of cellular processes.

Signaling Pathways necessary to create, and maintain MicroRNAs

The emergence of miRNA from scratch would involve a plethora of signaling pathways. These pathways play crucial roles in integrating various cellular stimuli and orchestrating specific responses. Here are some pivotal signaling pathways and their potential interconnections in relation to miRNA:

TGF-β/SMAD Pathway: Transforming growth factor-beta (TGF-β) is a crucial signaling molecule involved in various cellular processes. Activation of this pathway can lead to the transcription of specific miRNAs. SMAD proteins, integral components of this pathway, can bind directly to miRNA promoters, modulating their expression.
Wnt/β-catenin Pathway: The Wnt pathway is essential for many developmental processes. Upon pathway activation, stabilized β-catenin translocates to the nucleus and affects transcription. Certain miRNAs are direct targets of this pathway, and, conversely, some miRNAs can modulate the levels and activity of pathway components.
MAPK/ERK Pathway: The mitogen-activated protein kinase (MAPK) pathway is activated in response to various extracellular signals. Once activated, it can influence the expression of a variety of genes, including miRNAs. Some miRNAs target components of the MAPK pathway, forming feedback loops.
Notch Signaling: Activation of Notch receptors leads to the release of the Notch intracellular domain (NICD), which then moves to the nucleus and affects transcription. Notch signaling can induce or suppress the expression of specific miRNAs, which may then target components of the Notch pathway or other downstream effectors.
JAK-STAT Pathway: The Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is involved in responses to cytokines and growth factors. It can modulate miRNA expression, and certain miRNAs can in turn target and regulate pathway components.
PI3K/AKT/mTOR Pathway: This pathway is central to cellular growth and metabolism. It can regulate miRNA expression at multiple levels. Certain miRNAs, in return, target key components of this pathway, acting as regulators.

Interconnection, Interdependence, and Crosstalk

Feedback and Feedforward Loops: Many miRNAs target components of the signaling pathways that regulate their expression, forming intricate feedback or feedforward loops. These loops help maintain homeostasis and fine-tune responses.
Pathway Convergence: Multiple pathways might converge on the same miRNA, or a single pathway could regulate several miRNAs. This creates a web of interconnected regulations, allowing cells to integrate diverse signals and mount appropriate responses.
miRNA Sponges: Certain transcripts can act as "sponges" for miRNAs, sequestering them and preventing them from targeting their usual transcripts. This adds another layer to the interplay between miRNAs and signaling pathways.
Crosstalk with Other Systems: miRNAs don't only interact with signaling pathways. They're interconnected with the epigenetic machinery, metabolic pathways, and other post-transcriptional regulatory systems. For instance, changes in cellular metabolism might influence miRNA biogenesis, and epigenetic modifiers can regulate miRNA expression, adding depth to the regulatory network.

Given the vast complexity of signaling networks and their intersection with miRNA-mediated regulation, a coherent interplay of these systems is essential. Each piece of the network informs and is informed by multiple others, creating a harmonized cellular response to ever-changing conditions.

Regulatory codes necessary for maintenance and operation of MicroRNAs

miRNAs, in their operation and maintenance, would be subject to various regulatory codes and languages, which influence their biogenesis, stability, and function. These regulatory systems are intricate and precise, ensuring that miRNAs act at the right time, in the right place, and in response to the right cues. Promoter Sequences and Transcriptional Regulation: miRNA genes possess promoter regions just like protein-coding genes. These regions contain specific sequences recognized by transcription factors and RNA polymerase, which dictate when and where the miRNA is transcribed.

RNA Secondary Structures: The precursor miRNA forms specific secondary structures, like stem-loop structures, crucial for the recognition and processing by enzymes like Drosha in the nucleus.
Subcellular Localization Codes: After being processed in the nucleus, the precursor miRNA (pre-miRNA) is exported to the cytoplasm. This export is mediated by recognizing specific motifs on the pre-miRNA, ensuring efficient and timely transfer between cellular compartments.
Recognition Motifs for Enzymatic Processing: In the cytoplasm, pre-miRNAs are further processed into mature miRNAs by the enzyme Dicer. The recognition of pre-miRNAs by Dicer is also determined by specific structural motifs.
Seed Sequences: One of the defining features of miRNAs is their "seed sequence," a short region at the 5' end of the mature miRNA. This sequence drives the recognition and binding of the miRNA to target mRNAs. Its precise sequence and location within the miRNA are essential for target specificity.
RNA Modification Codes: miRNAs can be subjected to various modifications, such as methylation or uridylation. These modifications can influence miRNA stability, loading into the RNA-induced silencing complex (RISC), and efficiency in target repression.
Interactions with RNA-Binding Proteins (RBPs): Several RBPs can interact with miRNAs, influencing their stability, localization, or activity. These proteins often recognize specific motifs or structures in the miRNA or associated RNA molecules.
Feedback and Feedforward Loops: Many miRNAs are part of intricate regulatory loops that either repress or enhance the expression of factors that, in turn, regulate the miRNA's expression. This creates dynamic systems where miRNAs can rapidly respond to changes in their environment.
Integration with Cellular Stress Responses: In some instances, miRNAs play roles in cellular stress responses. They might be upregulated in response to specific stress signals and in turn modulate the expression of stress-related genes.
Temporal and Spatial Expression Patterns: The precise timing and location of miRNA expression are often crucial for their function. This is especially true during development, where the spatially and temporally controlled expression of miRNAs can influence cell fate decisions.

Understanding these codes and languages is fundamental to grasping the nuanced roles of miRNAs in cellular regulation. Each layer of control ensures that miRNAs can act as finely tuned regulators, integrating various signals to maintain cellular homeostasis and respond appropriately to changing environments.


Is there scientific evidence supporting the idea that microRNAs were brought about by the process of evolution?

miRNA systems present a striking intricacy in their design and function. This complexity, with multiple levels of interaction and control, suggests that a piecemeal or stepwise origin might face considerable challenges.

Precision of Interaction: miRNAs, in their mature form, must have specific sequences to accurately target messenger RNAs (mRNAs). A change or absence in this specific sequence would result in off-target effects or no binding at all. The emergence of an effective miRNA would require the coincidental formation of both the miRNA sequence and the target mRNA sequence, emphasizing a synchronized origin.
Biogenesis Dependency: The biogenesis of miRNA involves a series of coordinated steps, each mediated by specialized proteins and enzymes such as Drosha and Dicer. Without these proteins, precursor miRNAs wouldn't be processed into their mature forms, rendering them inactive. The concurrent evolution of both the miRNA sequences and the processing machinery seems a daunting task.
Regulation Complexity: miRNAs are not just passive entities but are subject to intricate regulation. This includes their transcription, processing, modifications, interactions with RNA-binding proteins, and incorporation into the RNA-induced silencing complex (RISC). Each of these steps is vital for the miRNA's activity and is controlled by a plethora of factors.
Feedback Mechanisms: Many miRNAs are part of complex feedback and feedforward loops, where they regulate and are regulated by other genes. This interconnected regulatory web suggests that the genes and the miRNAs co-evolved in a highly synchronized manner, making a stepwise evolution hard to envision.
Functional Redundancy: Several miRNAs can target the same mRNA or set of mRNAs. The redundancy might be seen as a buffer against perturbations, but it also raises questions about the evolutionary pressures that would maintain such overlapping functions.
Contextual Action: The action of miRNAs is highly context-dependent, meaning they might suppress a target in one tissue or developmental stage but not another. The emergence of such specificity would require coordinated changes in both the miRNA and the cellular context.

Given these factors, the miRNA system showcases features of intentional design rather than the result of gradual, unplanned processes. The concurrent existence of miRNAs, their specific targets, their processing machinery, and their regulatory systems hint at an integrated system set in place with forethought and precision. The potential pitfalls and inefficiencies in a stepwise evolutionary path for such an intricate system lead some to conclude that it bears the marks of deliberate orchestration.

Irreducibility and Interdependence of MicroRNAs to instantiate and operate

miRNA systems epitomize the intricate interplay of manufacturing, signaling, and regulatory codes and languages that govern cellular functions. Their presence and function raise fundamental questions about the origin and evolution of such a sophisticated system.

Irreducibility in Manufacturing: miRNA biogenesis involves a cascade of specific events that begin with the transcription of primary miRNA transcripts and culminate in the generation of mature miRNAs. This process involves precise protein machinery, such as the Drosha and Dicer enzymes. If any of these steps or components were missing or non-functional, the entire system would be rendered ineffective.
Interdependence in Signaling: Once formed, mature miRNAs don't act in isolation. They must be incorporated into the RNA-induced silencing complex (RISC). This complex, when equipped with the appropriate miRNA, then targets specific mRNAs for degradation or translational repression. The signaling is precise and requires both the miRNA and RISC components to be perfectly matched.
Regulatory Codes and Languages: The transcription and processing of miRNAs are not arbitrary but are subject to layers of regulation. Various transcription factors control the expression of miRNAs, while post-transcriptional modifications and interactions with RNA-binding proteins further refine their activity. These regulatory codes ensure that miRNAs act at the right place, at the right time, and in the right context.
Essential Communication Systems: Beyond their direct targets, miRNAs communicate with broader cellular pathways. They can influence, and be influenced by, signaling pathways, metabolic circuits, and stress responses. This crosstalk ensures that the cell's response is coordinated and fine-tuned.

Considering the intricacies of miRNA function and its dependencies, one is led to ponder how such a system might have originated. A partial or incomplete miRNA system seems to offer little advantage. Without the exact sequences, processing machinery, incorporation into RISC, and appropriate targets, miRNAs would not function as intended. A malfunctioning or imprecise miRNA system could be deleterious, leading to inappropriate gene silencing. Thus, the coordinated and interdependent nature of the miRNA system suggests it was introduced into biological systems fully formed and functional. The system's precision, its multifaceted interactions, and its essential role in cellular communication point to a design that is both intricate and deliberate.

Once is instantiated and operational, what other intra and extracellular systems are MicroRNAsinterdependent with?

miRNA, once instantiated and operational, forms an integral part of cellular function and exhibits a vast web of interactions within and outside the cell. Its interdependency with other systems is critical for orchestrating various cellular processes:

Transcriptional Machinery: miRNAs themselves are transcribed like other genes, often under the control of specific transcription factors. Thus, they are intimately connected with the cellular machinery that drives gene expression.
RNA Binding Proteins (RBPs): These proteins often modulate miRNA processing, stability, and function. They can interact with miRNAs and influence their maturation or activity.
RNA-induced silencing complex (RISC): After maturation, miRNAs are incorporated into the RISC, enabling them to exert their gene-silencing function. The components of RISC and miRNAs work synergistically to target and regulate specific mRNAs.
Endocytic Pathways: Some miRNAs and their associated proteins are shuttled inside endosomes and can even be expelled from the cell via exosomes, influencing neighboring or even distant cells.
Exosomes: These vesicles can contain miRNAs and can be secreted to the extracellular environment. Once released, exosomes can be taken up by other cells, transferring their miRNA content and influencing the recipient cell's functions.
Cell Signaling Networks: miRNAs play pivotal roles in a variety of signaling pathways, either as regulators or as outputs. For instance, they can be involved in response to growth factors, hormones, or stress signals.
Cell Cycle Machinery: miRNAs have roles in controlling cell proliferation, influencing different stages of the cell cycle, and ensuring timely progression or, in some cases, halting the cycle in response to stresses.
Apoptotic Pathways: Several miRNAs are known to either promote or inhibit apoptosis, thereby deciding the fate of the cell.
Stem Cell Maintenance and Differentiation: miRNAs are key regulators of stemness. They help maintain pluripotency in certain contexts and drive differentiation in others.
Extracellular Matrix (ECM): By modulating the expression of ECM components or enzymes that remodel the ECM, miRNAs can influence cell-matrix interactions, impacting processes like cell migration, tissue repair, and more.
Immune System: In immune cells, miRNAs play a vital role in modulating responses to pathogens, influencing differentiation, proliferation, and activation of various immune cell types.

The intricate interconnections between miRNAs and other cellular systems underscore their central role in cell biology. Their wide-ranging influences on cellular processes indicate that their regulatory potential was harnessed to fine-tune cellular responses to various internal and external cues, ensuring cellular and, ultimately, organismal homeostasis.

Major Premise: Systems that are based on semiotic codes and languages and exhibit profound interdependency are inherently complex and necessitate a level of coordination where each component is essential to the functioning of the whole.
Minor Premise: miRNAs and their associated pathways display this intricate semiotic coding, language-based regulation, and profound interdependency with multiple cellular systems.
Conclusion: Therefore, the emergence of miRNAs and their network of interactions suggests a coordinated, purposeful design, where each component and pathway had to be precisely instantiated to ensure the harmonious function of the overarching system.

https://reasonandscience.catsboard.com

Sponsored content



Back to top  Message [Page 1 of 1]

Permissions in this forum:
You cannot reply to topics in this forum