Fact-checked by Grok 2 weeks ago

Cell signaling

Cell signaling, also known as , is the fundamental process by which cells detect and respond to external stimuli from their or neighboring cells, enabling coordinated regulation of cellular activities such as , , , , and death. This communication is vital for all organisms, from single-celled that sense nutrient gradients to complex multicellular systems where it orchestrates , , and immune responses. In multicellular organisms, cell signaling occurs through diverse modes categorized by the distance and specificity of . Endocrine signaling involves hormones like insulin traveling through the bloodstream to distant target cells, regulating processes such as glucose across the body. employs molecules like growth factors that act locally on nearby cells, facilitating short-range coordination such as in or synaptic transmission. allows cells to respond to their own secreted signals, often playing roles in self-sustained , as seen with certain cancer cells producing their own growth factors. Additional modes include direct contact-dependent signaling via cell surface proteins and synaptic signaling in neurons using neurotransmitters like . Signaling molecules encompass a wide array, including hydrophilic peptides and proteins (e.g., ), hydrophobic (e.g., ), gases like , and lipids such as prostaglandins, each adapted to diffuse or bind specific receptors. Receptors, which bind these with high affinity (often at concentrations below 10^{-8} M), are primarily transmembrane proteins on the cell surface for water-soluble signals or intracellular for lipophilic ones like steroid hormones that directly influence . Upon ligand binding, receptors initiate intracellular pathways, frequently involving second messengers such as cyclic AMP or calcium ions, which amplify the signal and activate cascades of protein kinases to elicit diverse cellular responses. Dysregulation of cell signaling pathways underlies numerous diseases, including cancer—where mutations in signaling components like receptor tyrosine kinases promote uncontrolled growth—and , highlighting the therapeutic potential of targeting these pathways. Ongoing research continues to uncover intricate networks, such as those integrating multiple signals for context-specific outcomes, underscoring cell signaling's role as a cornerstone of cellular and organismal .

Overview

Definition and scope

Cell signaling is the process by which cells detect and respond to internal and external molecular cues, enabling communication between cells and coordination of physiological functions across organisms. This involves the transmission of signals from a sending cell or the to a receiving cell, typically through ligands—diverse molecules such as hormones, neurotransmitters, or growth factors—that bind to specific receptors on or within the target cell, triggering intracellular changes that alter cellular . The of cell signaling encompasses both unicellular and multicellular organisms, where it facilitates responses to stimuli like nutrients, pathogens, or mechanical stress, ensuring survival, growth, and adaptation without requiring direct physical contact. At its core, cell signaling relies on four key components: ligands, which serve as the primary signaling molecules; receptors, which are specialized proteins that recognize and bind ligands with high ; second messengers, such as cyclic AMP (), inositol (IP₃), or calcium ions (Ca²⁺), that propagate and amplify the signal inside the cell; and effectors, including enzymes like kinases or transcription factors, that execute the final cellular outcomes. These elements form integrated networks that allow precise regulation, where even low concentrations of extracellular signals can elicit robust intracellular responses through cascading amplification. The fundamental process of cell signaling unfolds in three stages: , where binding induces a conformational change in the receptor; , involving relay mechanisms like enzymatic activations or second messenger that convert the external signal into intracellular events; and response, where the amplified signal drives specific outcomes such as cytoskeletal reorganization or changes. This streamlined pathway ensures efficient information transfer while minimizing noise, allowing cells to integrate multiple signals for context-dependent decisions. Cell signaling is indispensable for enabling cell differentiation, proliferation, apoptosis, and environmental adaptation, playing pivotal roles in embryonic development, activation, and tissue repair processes that restore after injury. Dysregulation of these pathways underlies diseases like cancer and , highlighting their broad biological impact. Historically, the field built on Earl W. Sutherland's pioneering 1950s experiments demonstrating as the first intracellular second messenger in hormone action, for which he received the 1971 Nobel Prize in or ; the modern term "cell signaling" emerged in the late , formalizing concepts from the 1970s era of research.

Evolutionary and taxonomic aspects

Cell signaling mechanisms originated in prokaryotes, with early forms evident in bacterial quorum sensing systems that utilize autoinducers to coordinate population-level behaviors such as biofilm formation and virulence factor expression. These pathways, which rely on diffusible signaling molecules to detect cell density, represent an ancient adaptation for environmental sensing and survival, predating the divergence of major bacterial lineages. In parallel, two-component systems in bacteria, involving histidine kinases and response regulators, emerged as a foundational mode of signal transduction for detecting nutrients, toxins, and osmotic changes, enabling rapid adaptive responses. These prokaryotic signaling strategies are conserved and expanded in eukaryotes, from unicellular organisms like to complex multicellular animals, reflecting a shared evolutionary heritage. Core elements, such as receptor-ligand binding and cascades, trace back to prokaryotic ancestors and have been reiterated across domains. For instance, two-component systems in prokaryotes and (MAPK) cascades in eukaryotes exemplify modular signal amplification in diverse contexts. In eukaryotes, these mechanisms underpin developmental processes, with expansions in metazoans allowing for tissue-specific coordination, as seen in the integration of signaling modules for . Cell signaling is ubiquitous across all domains of life, manifesting in through chemotaxis pathways that direct motility toward favorable conditions via two-component-like systems distinct from bacterial homologs yet functionally analogous. In , two-component systems dominate, facilitating environmental adaptation, while in Eukarya, G-protein-coupled receptors (GPCRs) are prevalent in for hormone perception, in fungi for nutrient and stress responses, and in for sensory and intercellular communication. Prokaryotic signaling primarily supports survival strategies like nutrient sensing and population coordination, whereas eukaryotic adaptations emphasize developmental patterning, exemplified by the Wnt pathway's role in axis formation and tissue morphogenesis in . Horizontal gene transfer has significantly contributed to the dissemination of signaling genes across taxa, particularly two-component systems that originated in and spread to and eukaryotes, enhancing adaptive versatility in recipient organisms. This process underscores the dynamic evolution of signaling networks, allowing prokaryotes to acquire eukaryotic-like modules and vice versa, thereby blurring strict domain boundaries in architecture.

Signaling Molecules

Extracellular signals

Extracellular signals are molecules produced by cells and released into the extracellular environment to communicate with other cells, enabling coordinated responses in multicellular organisms. These signals traverse the , interstitial fluids, or bloodstream, with their range determined by factors such as molecular size, solubility, and degradation rates—local signals like growth factors act over short distances (micrometers to millimeters), while systemic ones like hormones can travel throughout the body. The synthesis and release of these signals are tightly regulated to maintain physiological , and their chemical diversity allows for specific interactions in various biological contexts. Extracellular signals are broadly classified by their chemical nature and function. Peptide and protein signals, such as hormones (e.g., insulin), cytokines (e.g., ), and growth factors (e.g., ), are hydrophilic and typically water-soluble, requiring vesicular transport for release. Small-molecule neurotransmitters, such as , are also hydrophilic but not peptide-based. In contrast, steroid hormones (e.g., ) are lipophilic, derived from , and diffuse across membranes. Lipid-derived signals, such as prostaglandins and other eicosanoids, are hydrophobic molecules synthesized from released from membrane phospholipids via enzymes like (). Gaseous signals like (NO) are small, non-polar molecules produced on demand without storage. This classification influences their stability and mode of action, with peptides often having short half-lives (minutes) due to enzymatic degradation, while steroids persist longer (hours). Synthesis of extracellular signals occurs in specialized cellular compartments. Peptide signals are transcribed from genes, translated in the rough (), and processed through glycosylation and cleavage in the Golgi apparatus before packaging into secretory vesicles. Steroid signals are synthesized in the smooth and mitochondria of endocrine cells, involving enzymatic conversions of cholesterol via enzymes. Lipid-derived signals like prostaglandins are produced in the and membranes through the pathway. Gasotransmitters like NO are generated enzymatically by () in the , using L-arginine as a substrate, with production regulated by calcium-calmodulin binding. Transcriptional control, such as via nuclear receptors, and post-translational modifications like ensure precise regulation of signal production in response to cellular cues. Release mechanisms primarily involve exocytosis, where signaling molecules in vesicles fuse with the plasma membrane in a calcium-dependent manner, triggered by depolarization or receptor activation. For instance, insulin is released from pancreatic beta cells via regulated exocytosis following glucose-stimulated insulin secretion. Small lipophilic molecules like steroids, lipids, and gases such as NO are released by simple diffusion across the lipid bilayer, bypassing vesicular pathways. Unconventional secretion, including direct translocation or extracellular vesicle release, applies to certain cytokines and growth factors that lack signal peptides. Concentration gradients form post-release, with half-lives varying—acetylcholine is rapidly hydrolyzed by acetylcholinesterase (milliseconds), ensuring transient synaptic signaling, while insulin circulates with a half-life of about 5-10 minutes. These processes allow extracellular signals to mediate both local autocrine/paracrine effects and distant endocrine communication.

Intracellular signals

Intracellular signals, commonly known as second messengers, are small molecules or ions produced within the cell in response to the activation of cell surface receptors by extracellular ligands. These include (cAMP), inositol 1,4,5-trisphosphate (IP₃), diacylglycerol (DAG), and calcium ions (Ca²⁺), which serve to transduce and amplify the initial signal inside the cell. The generation of these second messengers typically occurs through receptor-coupled enzymes that catalyze rapid biochemical reactions. For instance, in (GPCR) pathways, activation of converts ATP to , while (PLC) hydrolyzes (PIP₂) in the plasma membrane to produce IP₃ and DAG. Enzymatic cascades further amplify these signals, allowing a single receptor activation to generate thousands of second messenger molecules. Ca²⁺, often mobilized from intracellular stores like the in response to IP₃ binding to its receptors, also acts as a key second messenger. These molecules relay information from the plasma membrane to intracellular targets, such as enzymes in the or transcription factors in the , enabling diverse cellular responses. Spatial and temporal control is achieved through gradients, binding to proteins, or localized production, which prevents indiscriminate signaling. For example, cAMP in GPCR pathways binds to and activates (PKA), leading to of downstream targets that regulate processes like glycogen breakdown. Similarly, Ca²⁺ waves propagate across cells to trigger muscle contraction in cardiomyocytes or synaptic vesicle secretion in neurons. Signal termination is critical for specificity; degradation mechanisms, such as cAMP phosphodiesterases converting cAMP to AMP, rapidly lower concentrations, often restoring levels from stimulated micromolar ranges (e.g., 1–10 μM for Ca²⁺ or cAMP peaks) back to basal nanomolar states.

Modes of Intercellular Communication

Autocrine and paracrine signaling

refers to a mode of cellular communication in which a produces a signaling molecule, or , that binds to receptors on its own surface, thereby influencing its own without involving other cells. This self-regulatory mechanism allows cells to fine-tune their responses to internal or environmental cues, often amplifying or sustaining specific physiological processes. In contrast, involves the release of ligands that diffuse through the to act on neighboring cells within a localized area, enabling coordinated responses among adjacent cells without systemic spread. Both forms represent short-range intercellular communication, distinguished from longer-range endocrine signaling by their reliance on over limited distances. In , the ligand's action on the producing can create feedback loops that promote cell survival, , or . A prominent example is the role of transforming growth factor-β (TGF-β) in cancer cells, where autocrine secretion of TGF-β binds to receptors on the same tumor cells, driving epithelial-to-mesenchymal transition and enhancing invasiveness. This autocrine loop sustains tumor growth by maintaining a pro- state, as observed in various carcinomas where TGF-β expression correlates with aggressive phenotypes. Such loops are particularly implicated in pathological conditions, where dysregulated autocrine signaling can perpetuate uncontrolled and resistance to . Paracrine signaling facilitates rapid, localized interactions essential for homeostasis and response to . For instance, in , platelets release (PDGF) that diffuses to nearby fibroblasts and endothelial cells, stimulating and to promote repair. Similarly, in immune , T cells secrete interleukin-2 (IL-2), which acts paracrine on adjacent lymphocytes to enhance and effector functions during inflammatory responses. Neurotransmitters at synapses exemplify in the , where molecules like glutamate diffuse across the synaptic cleft to activate receptors on postsynaptic cells, enabling swift . The mechanisms underlying both autocrine and paracrine signaling are governed by diffusion of soluble ligands through the , limiting their effective range to nanometers (e.g., synaptic clefts of ~20-40 ) up to a few millimeters in denser tissues. This short-range propagation ensures rapid onset of responses, often within seconds to minutes, followed by quick decay due to ligand degradation, uptake, or dilution, preventing unintended widespread effects. Ligand-receptor binding initiates downstream events via cell surface or intracellular receptors, though the core distinction lies in the spatial confinement of these interactions. In pathological contexts, such as tumor microenvironments, autocrine loops involving growth factors like TGF-β can drive autonomous cancer progression, underscoring their role in disease while highlighting their separation from distant endocrine modes by proximity.

Endocrine and juxtacrine signaling

Endocrine signaling involves the secretion of hormones by specialized endocrine cells, which are then transported through the bloodstream to act on distant target cells throughout the body. This mode of communication enables systemic regulation of physiological processes, such as , , and stress responses. For instance, adrenaline (epinephrine), released from the adrenal glands, circulates via the blood to bind β-adrenergic receptors on target cells like those in the heart and lungs, triggering the by increasing heart rate and bronchodilation. Similarly, , produced by the gland, travel through the circulation to regulate by binding to nuclear receptors in various tissues, thereby influencing energy expenditure and . In contrast, juxtacrine signaling requires direct physical contact between , typically mediated by membrane-bound on one cell interacting with receptors on an adjacent cell. This contact-dependent mechanism ensures highly localized and precise signal transmission, often crucial for developmental processes like and tissue patterning. A prominent example is the Notch-Delta pathway, where the membrane-bound ligand Delta on a signaling cell binds the receptor on a neighboring cell, leading to proteolytic of Notch and release of its intracellular to modulate for decisions in cell during embryogenesis. Another key instance is Ephrin-Eph signaling, where membrane-anchored ephrins bind Eph receptors on adjacent cells to provide bidirectional cues that guide in the , preventing misguided projections and establishing topographic maps. The mechanisms underlying endocrine signaling rely on the for hormone distribution, with target specificity achieved through selective binding to cell surface or intracellular receptors that recognize particular . often exhibit longer half-lives in the blood—ranging from minutes for catecholamines like adrenaline to hours or days for and —allowing sustained effects but potentially lower spatial specificity compared to local signals like paracrine . Juxtacrine signaling, however, utilizes molecules and transmembrane interactions for signal transfer without diffusible mediators, enabling rapid, high-fidelity patterning in tissues where precise - coordination is essential, such as in boundary formation during . This contact-based nature confers greater specificity and immediacy, contrasting with the broader reach of endocrine signals.

Receptors

Cell surface receptors

Cell surface receptors are membrane proteins embedded in the that detect extracellular signaling molecules, primarily hydrophilic ligands unable to cross the , and transduce these signals into intracellular responses. These receptors typically consist of an extracellular ligand-binding domain that recognizes specific signals, a hydrophobic that spans the bilayer, and an intracellular signaling domain that interacts with downstream effectors to propagate the signal. Unlike intracellular receptors, which bind lipophilic ligands in the or , cell surface receptors serve as the primary interface for cell-cell communication via water-soluble messengers. The major classes of cell surface receptors are distinguished by their structure and mechanism of action. Ion channel-linked receptors, also known as ligand-gated ion channels, form pores that open or close upon binding, enabling rapid changes in through ion flux such as Na⁺, K⁺, Ca²⁺, or Cl⁻. A representative example is the , which binds to allow sodium influx, facilitating synaptic transmission in neuromuscular junctions. G protein-coupled receptors (GPCRs) represent the largest superfamily, comprising approximately 800 genes in the and accounting for about 4% of protein-coding genes. These receptors feature seven α-helical transmembrane domains that form a barrel-like structure, with an extracellular for ligand recognition and an intracellular that couples to heterotrimeric G proteins, activating pathways involving second messengers like cyclic AMP () or inositol trisphosphate (). GPCRs detect diverse stimuli, including light through in rod cells of the and odors via olfactory receptors in nasal . Enzyme-linked receptors contain or associate with enzymatic domains that catalyze or other modifications upon . Receptor tyrosine kinases (RTKs), a key subclass, dimerize upon ligand binding, leading to autophosphorylation on intracellular residues and recruitment of signaling proteins. The (EGFR), for instance, binds (EGF) to initiate cascades promoting and survival. Ligand binding to cell surface receptors exhibits high specificity, often following the lock-and-key model where the precisely fits a preformed binding pocket, or the induced fit model involving conformational changes in the receptor to optimize interactions. Binding affinity is quantified by the (K_d), typically in the nanomolar (nM) range (e.g., 0.3–0.5 nM for certain receptor-ligand pairs), ensuring sensitive detection of low-concentration signals. Mutations in receptor genes underlie numerous diseases, including from insulin receptor defects and certain cancers from EGFR dysregulation, highlighting their clinical significance.

Intracellular receptors

Intracellular receptors, primarily comprising the superfamily, are ligand-activated transcription factors located within the cell, either in the or , that respond to lipophilic signaling molecules capable of crossing the plasma membrane. These receptors include receptors, such as the (ER), (AR), glucocorticoid receptor (GR), progesterone receptor (PR), and mineralocorticoid receptor (MR); non-steroid receptors like thyroid hormone receptor (TR), retinoic acid receptor (RAR), vitamin D receptor (VDR), and peroxisome proliferator-activated receptor (PPAR); and orphan receptors, such as the testicular receptor, for which endogenous ligands remain unidentified. In humans, approximately 48 s have been identified, reflecting their diverse roles in regulating cellular processes like and . The mechanism of action for these receptors involves the passive diffusion of lipophilic ligands—such as steroids, thyroid hormones, retinoic acid, and vitamin D—across the lipid bilayer of the cell membrane. Upon entering the cell, the ligand binds to the receptor, inducing a conformational change that often releases inhibitory chaperone proteins like heat shock proteins from cytosolic receptors (e.g., GR), exposing a nuclear localization signal that facilitates translocation to the nucleus. The ligand-receptor complex then binds to specific DNA sequences known as hormone response elements (HREs) as homodimers, heterodimers, or monomers, recruiting co-activators or co-repressors to modulate target gene transcription, thereby directly influencing gene expression without intermediary second messengers. Structurally, receptors share a conserved modular , including an N-terminal (NTD) for , a central (DBD) featuring two motifs that recognize HREs, a flexible region containing the localization signal, and a C-terminal -binding (LBD) with a hydrophobic pocket for accommodation and an function-2 (AF-2) that interacts with co-activators or co-repressors to regulate transcriptional activity. These structural elements enable precise specificity and DNA targeting, with co-regulators fine-tuning the receptor's repressive or activating effects on . A prominent example is the (), which, upon binding glucocorticoids, translocates to the and inhibits pro-inflammatory genes by interfering with transcription factors like , thereby suppressing inflammation. Another key instance involves (RXR) heterodimers, such as RXR-PPAR or RXR-LXR complexes, which regulate by activating genes involved in oxidation, efflux, and in the liver. Evolutionarily, nuclear receptors trace back to ancient lipid sensors that emerged around 600 million years ago in bilaterian ancestors, initially functioning as promiscuous detectors of and sterols before diversifying into specialized ligand-responsive transcription factors.

Signal Transduction Pathways

Initiation and amplification

Cell signaling begins with the binding of an extracellular to a specific receptor, which induces a conformational change in the receptor protein, thereby activating downstream effectors. This initial event translates the extracellular signal into an intracellular response, often involving the recruitment and activation of adapter proteins or enzymes that propagate the signal. For instance, in G protein-coupled receptors (GPCRs), ligand binding stabilizes an active receptor conformation that facilitates the interaction with heterotrimeric s, promoting the release of (GDP) from the Gα subunit and its exchange for (GTP). This GDP-GTP exchange dissociates the G protein into active Gα-GTP and Gβγ subunits, each capable of modulating effector enzymes. Signal amplification occurs through multistep enzymatic cascades and the generation of second messengers, allowing a single activated receptor to elicit a robust cellular response. In these cascades, each activated molecule can catalyze the activation of numerous downstream targets, exponentially increasing the signal strength. For example, activated Gαs stimulates to produce hundreds of () molecules per second, which in turn activate multiple () holoenzymes; each PKA can phosphorylate dozens of substrates, further propagating the signal. Similarly, in the (PLC) pathway, activated PLC hydrolyzes (PIP₂) into inositol 1,4,5-trisphosphate (IP₃) and diacylglycerol (DAG), with each enzyme molecule generating multiple second messengers that mobilize intracellular calcium and activate (PKC), respectively. Phosphorylation-dephosphorylation cycles contribute to this amplification by enabling ultrasensitive switches, where kinase-phosphatase pairs convert low-level inputs into sharp, all-or-nothing outputs through mechanisms like substrate and multistep binding, achieving Hill coefficients up to n+1 (where n is the number of phosphorylation sites). Second messengers such as and IP₃ diffuse rapidly within the , spreading the signal to distal effectors and enhancing amplification. To ensure specificity and prevent crosstalk between pathways, is spatially regulated by scaffold proteins that organize signaling components into localized complexes. These scaffolds tether receptors, kinases, and second messengers, confining signals to microdomains and limiting diffusion-based interference; for example, A-kinase anchoring proteins (AKAPs) localize near in the pathway. Overall, such mechanisms allow cells to detect and respond effectively to trace concentrations, with ratios potentially exceeding 10⁴ in kinase cascades like MAPK, though exact gains vary by pathway.

Major signaling cascades

Cell signaling pathways often converge on a set of conserved cascades that transduce extracellular cues into intracellular responses, enabling cells to coordinate processes like , , and . These major signaling cascades, including the cAMP-PKA, MAPK/ERK, PI3K-Akt, , Wnt, and TGF-β pathways, exhibit modular architectures where receptor activation triggers sequential enzymatic activations leading to effector modulation. Other prominent cascades include the JAK- pathway, activated by receptors to promote immune responses and hematopoiesis via STAT transcription factors, and the pathway, which regulates and cell through IκB degradation and nuclear translocation of NF-κB. While amplification mechanisms, such as second messenger production, enhance signal strength within these cascades, their specificity arises from scaffold proteins and compartmentalization. The cAMP-PKA pathway is activated by G protein-coupled receptors (GPCRs) coupled to stimulatory G proteins (Gs), which upon ligand binding stimulate to produce cyclic AMP () from ATP. Elevated levels activate (PKA) by binding to its regulatory subunits, releasing catalytic subunits that phosphorylate diverse targets, including the transcription factor CREB to regulate . This pathway is pivotal in responses to hormones like adrenaline, influencing and activity. The MAPK/ERK pathway, often initiated by receptor tyrosine kinases (RTKs), propagates signals through a kinase cascade: ligand-bound RTKs recruit and activate , which in turn activates , leading to sequential and activation of MEK and then ERK. Activated ERK translocates to the to phosphorylate transcription factors, promoting and in response to growth factors like EGF. Non-canonical branches, such as the JNK pathway, diverge from this core to handle stress signals, where JNK activation occurs via MAP3K like ASK1 in response to UV or cytokines. The PI3K-Akt pathway is triggered by RTKs or GPCRs, recruiting (PI3K) to the membrane, where it phosphorylates PIP2 to generate PIP3, a second messenger that recruits and activates Akt kinase via PDK1 phosphorylation. Akt then phosphorylates downstream effectors like , regulating cell survival, metabolism, and protein synthesis; dysregulation, such as PTEN mutations that inactivate the PIP3 , hyperactivates this pathway and drives oncogenesis in cancers like . The pathway operates via juxtacrine signaling, where binding on an adjacent cell induces sequential proteolytic cleavages of the receptor by metalloproteases and γ-secretase, releasing the intracellular domain (NICD) that translocates to the nucleus to co-activate transcription factors like CSL, influencing developmental decisions such as cell fate binary choices. Recent studies highlight 's ongoing role in maintenance, including asymmetric division in intestinal stem cells and regeneration in muscle satellite cells. Other prominent cascades include the Wnt pathway, where Wnt ligands bind receptors to inhibit the β-catenin destruction complex (APC/Axin/GSK3/CK1), stabilizing β-catenin for nuclear translocation and TCF/LEF-mediated transcription of genes involved in embryogenesis and tissue . The TGF-β pathway, activated by TGF-β family ligands binding type I/II serine/threonine kinase receptors, leads to phosphorylation of receptor-regulated SMADs (R-SMADs) that complex with SMAD4 to enter the nucleus and regulate transcription of extracellular matrix genes critical for . These cascades do not function in isolation; cross-talk integrates multiple inputs, as seen in the convergence of MAPK and PI3K pathways on shared effectors like FoxO transcription factors to determine cell fate outcomes in versus decisions. Computational models now simulate these , incorporating kinetic parameters to predict pathway behaviors under varying stimuli and reveal emergent properties like . Recent advances as of 2025 include AI-driven modeling of pathway cross-talk and novel targeted therapies for diseases like cancer.

Cellular Responses

Immediate and short-term effects

Immediate and short-term effects of cell signaling encompass rapid cellular responses that occur without involving changes in , typically manifesting within seconds to minutes through alterations in fluxes, metabolic activities, and cytoskeletal dynamics. These effects enable quick adaptations to environmental cues, such as release or binding, by directly modulating existing cellular machinery. For instance, binding to receptors can trigger the opening of channels, leading to immediate changes in and concentrations that propagate signals across cells. One prominent type of immediate response is the opening of ion channels, exemplified by in neurons upon of glutamate receptors. Glutamate binding to ionotropic receptors, such as and NMDA types, opens cation-permeable channels, allowing influx of Na⁺ and Ca²⁺ ions, which rapidly the postsynaptic membrane and facilitates synaptic transmission. This process occurs on a millisecond timescale, enabling fast excitatory signaling in the . Similarly, cytoskeletal rearrangements represent another key short-term effect, driven by Rho GTPases that regulate dynamics for cell motility. of RhoA, for example, promotes formation and contractility, while Rac1 and Cdc42 induce lamellipodia and extension, respectively, allowing cells to migrate in response to chemotactic signals within minutes. Illustrative examples highlight the physiological relevance of these responses. In insulin signaling, receptor activation leads to the translocation of glucose transporters from intracellular vesicles to the plasma membrane, enhancing in adipocytes and muscle cells within minutes to support metabolic homeostasis. Likewise, Ca²⁺ release from intracellular stores, often triggered by (IP3), induces by binding to , which exposes actin-myosin binding sites, or promotes in secretory cells, such as release from synaptic vesicles. These effects are mediated by second messengers that amplify signals and activate effectors. Cyclic AMP (cAMP), generated by upon G-protein-coupled receptor activation, binds to (PKA), which phosphorylates ion channels and enzymes to alter their activity, such as opening cAMP-gated channels in sensory neurons. IP3, produced by , releases Ca²⁺ from the , which in turn activates calmodulin-dependent kinases to phosphorylate targets involved in or . Such mechanisms ensure responses are confined to seconds to minutes, allowing precise and transient cellular adjustments. Physiologically, these rapid effects underpin critical processes like synaptic transmission for neural communication, hormone-induced in endocrine cells, and acute responses that mobilize reserves. For example, epinephrine signaling via rapidly increases in liver cells to elevate blood glucose during fight-or-flight scenarios. These responses are inherently reversible and energy-efficient, relying on post-translational modifications rather than protein synthesis, which facilitates quick recovery. Disruptions in these pathways, such as mutations in ion channels leading to channelopathies, can result in disorders like , where aberrant channel opening causes hyperexcitability and seizures in neurons.

Long-term gene regulation

Cell signaling pathways often culminate in long-term alterations to gene expression, enabling sustained cellular adaptations such as differentiation and proliferation. These changes are primarily mediated through the activation and nuclear translocation of transcription factors, which bind to specific DNA sequences to regulate target genes over hours to days. For instance, in the mitogen-activated protein kinase (MAPK) pathway, extracellular signal-regulated kinase (ERK) phosphorylates the transcription factor Elk-1, promoting its activation and subsequent induction of immediate early genes like Fos and Egr1. Similarly, in the nuclear factor kappa B (NF-κB) pathway, signaling triggers the phosphorylation and ubiquitin-mediated degradation of the inhibitor IκB, releasing NF-κB dimers for nuclear translocation and binding to κB sites in promoters of genes involved in inflammation and survival. Steroid hormone receptors exemplify direct genomic regulation, where ligand binding induces conformational changes, nuclear localization, and dimerization, allowing the receptor-hormone complex to bind hormone response elements (HREs) on DNA and recruit co-activators to initiate transcription of target genes. In cytokine signaling, the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway facilitates rapid nuclear entry of phosphorylated STAT dimers, which bind interferon-stimulated response elements (ISREs) to activate hundreds of interferon-stimulated genes (ISGs) critical for antiviral defense and immune modulation. These transcription factors often interface with chromatin remodeling processes to facilitate enhancer activation and epigenetic modifications, ensuring stable gene expression changes. Chromatin remodelers, such as complexes, reposition nucleosomes at enhancers in response to signaling cues, increasing accessibility for transcription factor binding and promoting enhancer-promoter looping. Epigenetic mechanisms, including histone acetylation by co-activators like (CBP) and p300, further enhance this by neutralizing chromatin charge and recruiting additional factors to poised regulatory elements. The resulting gene expression programs drive profound outcomes, including shifts in cell fate; for example, , a myogenic regulatory factor activated downstream of signaling pathways like Wnt, binds enhancers to orchestrate the transcriptional network for differentiation during . These long-term effects, lasting from hours to days, contrast with transient metabolic responses by embedding persistent epigenetic marks that maintain altered cellular identity. Long-term gene regulation integrates inputs from multiple signaling pathways to fine-tune developmental processes, such as the interplay between and Wnt in embryogenesis, where Notch intracellular domain suppresses Wnt targets while co-regulating for somitogenesis and tissue patterning. Recent advances in single-cell sequencing (scRNA-seq) have illuminated these pathway-gene linkages at cellular resolution, revealing heterogeneous responses and novel regulatory networks in contexts like immune activation and development since 2015.

Regulation and Modulation

Receptor desensitization

Receptor desensitization refers to the adaptive processes that attenuate cellular responsiveness to persistent or repeated stimulation, thereby preventing excessive signaling and maintaining . This phenomenon occurs at multiple levels, including rapid uncoupling of receptors from downstream effectors and longer-term reduction in receptor availability through trafficking. These mechanisms are crucial for modulating signal duration and intensity across various receptor families, such as G protein-coupled receptors (GPCRs) and (RTKs). Short-term desensitization primarily involves post-translational modifications that inactivate receptors within seconds to minutes. For GPCRs, agonist binding induces phosphorylation of serine and threonine residues in the receptor's cytoplasmic domains by G protein-coupled receptor kinases (GRKs), such as GRK2 and GRK3. This phosphorylation recruits β-arrestins (e.g., β-arrestin1 and β-arrestin2), which bind to the phosphorylated receptor and sterically hinder G protein coupling, thereby uncoupling the receptor from its signaling partners and terminating G protein-mediated responses like cAMP production. β-Arrestins not only block signaling but also serve as scaffolds for additional regulatory proteins, further fine-tuning the response. In RTKs, analogous rapid inactivation can occur through autophosphorylation followed by recruitment of inhibitory phosphatases or adaptors, though the emphasis here is on subsequent trafficking events. These acute processes protect cells from overstimulation, as unchecked activation could lead to toxicity or pathological states. A key mechanism for longer-term desensitization is receptor internalization via endocytosis, which removes receptors from the cell surface and either recycles them or directs them to degradation. In GPCRs, β-arrestin-bound receptors are internalized through clathrin-coated pits, involving adaptor proteins like AP-2, leading to sequestration in endosomes. Depending on the receptor and ligand, internalized GPCRs may recycle to the plasma membrane to restore sensitivity or be sorted to lysosomes for degradation, balancing responsiveness. For RTKs, such as the epidermal growth factor receptor (EGFR), ligand-induced dimerization triggers ubiquitination by E3 ligases like c-Cbl, marking the receptor for clathrin-mediated endocytosis. Ubiquitinated RTKs are then trafficked through early endosomes to multivesicular bodies and ultimately to lysosomes, where degradation attenuates signaling and downregulates surface receptor levels. This recycling versus degradation dichotomy allows cells to adapt sensitivity based on stimulus duration. Illustrative examples highlight the physiological and pathological implications of desensitization. In opioid tolerance, chronic exposure to agonists like induces μ-opioid receptor () phosphorylation by GRK2/3, followed by β-arrestin recruitment and , reducing analgesic efficacy and contributing to —a rapid loss of response to repeated stimulation. Similarly, in cancer, impaired and , or compensatory pathway , can contribute to resistance against targeted therapies like , sustaining oncogenic signaling despite initial promotion of . Recent models incorporating biased agonism, where ligands preferentially activate or β-arrestin pathways, have refined our understanding since the 2010s, revealing how pathway-specific desensitization influences therapeutic outcomes without uniform attenuation. Overall, these processes prevent overstimulation during chronic exposure, with emerging as a hallmark of adaptive desensitization.

Feedback mechanisms

Feedback mechanisms in cell signaling are system-level controls that maintain by fine-tuning the activity of signaling networks through and loops. These loops enable cells to respond adaptively to stimuli, preventing overactivation or insufficient signaling, and contribute to the robustness of biological systems. typically inhibits pathway components to terminate or attenuate signals, while amplifies responses, often creating bistable switches for decisive cellular decisions such as . Negative feedback loops inhibit signaling pathways to prevent excessive activation and promote signal termination. In insulin signaling, Akt and its downstream effectors, such as S6K1, phosphorylate substrate-1 (IRS-1) on serine residues, which dampens IRS-1's ability to recruit downstream effectors and thus attenuates the pathway. Similarly, protein phosphatase 2A (PP2A) acts as a negative in the ERK/MAPK cascade by dephosphorylating activated ERK1/2 and MEK1/2, thereby reducing signal propagation and restoring basal states. Positive feedback loops amplify signals to generate switch-like behaviors, facilitating rapid transitions in cellular states. In the MAPK pathway, Ras-GTP enhances the (GEF) activity of SOS, which further activates Ras, creating a bistable switch that supports processes, such as in PC-12 cells responding to EGF. Network integration occurs through crosstalk between pathways and organization by scaffold proteins, allowing coordinated regulation via . For instance, PI3K/AKT signaling inhibits GSK3β through Ser9 , stabilizing β-catenin and enhancing Wnt pathway activity, which promotes transcriptional responses like EMT in cancer contexts. Scaffold proteins, such as A-kinase anchoring proteins (AKAPs), organize kinases and phosphatases into complexes, recruiting termination enzymes like PP2A to enforce and modulate network dynamics. Representative examples illustrate these mechanisms in broader physiological contexts. In circadian rhythms, the PER/CRY complex forms a loop by rhythmically inhibiting CLOCK:BMAL1 transcriptional activity, with PER2 serving as a scaffold to bridge CRY and repressors, ensuring oscillatory essential for daily cycles. In immune tolerance, CTLA-4 on regulatory T cells provides by competing with for / ligands on antigen-presenting cells, dampening T cell activation and preventing . These feedback mechanisms ensure signaling robustness by buffering perturbations and maintaining steady states. Dysregulation of feedback loops contributes to diseases; for example, dysregulated positive feedback loops in Th17 cell signaling, such as IL-17-induced IL-6 production amplifying via /, underlie conditions like and experimental autoimmune encephalomyelitis. approaches, including dynamic differential equation models of gene regulatory networks, quantify loop strengths and interactions in the 2020s, enabling predictions of signaling outcomes in and .

References

  1. [1]
    Cell Signaling - The Cell - NCBI Bookshelf - NIH
    Chapter 13Cell Signaling. All cells receive and respond to signals from their surroundings. Even the simplest bacteria sense and swim toward high concentrations ...
  2. [2]
    Signaling Molecules and Their Receptors - The Cell - NCBI Bookshelf
    Cell signaling can take place either through direct cell-cell contacts or through the action of secreted signaling molecules. (A) In endocrine signaling, ...Modes of Cell-Cell Signaling · Steroid Hormones and the... · Neurotransmitters
  3. [3]
    General Principles of Cell Communication - NCBI - NIH
    Cell signaling requires not only extracellular signal molecules, but also a complementary set of receptor proteins in each cell that enable it to bind and ...
  4. [4]
    Cell Signaling | Learn Science at Scitable - Nature
    Cells receive chemical signals, often via receptors, which trigger a chain of events that amplifies the signal inside the cell. Cells also send signals.Missing: key | Show results with:key
  5. [5]
    Conceptual Evolution of Cell Signaling - PMC - PubMed Central - NIH
    The cellular signaling starts as soon as the first messenger (the ligand) binds to its receptor—a protein with the complementary structure on a transmembrane ...
  6. [6]
    Cell–cell communication: new insights and clinical implications
    Aug 7, 2024 · Cell signaling, which involves transmitting information between cells or subcellular components, is an inherent characteristic of living ...
  7. [7]
    [PDF] cell signaling - UC Berkeley MCB
    Cell signaling involves detecting stimuli, transferring signals to the cytoplasm, and transmitting them to effector molecules, causing a cellular response. ...
  8. [8]
    Second Messengers - PMC - NIH
    Second messengers are small molecules and ions that relay signals received by cell-surface receptors to effector proteins.
  9. [9]
    Cell Signaling - an overview | ScienceDirect Topics
    They are known for their immunomodulatory and pro-regenerative signaling capabilities, playing a crucial role in tissue repair and regeneration. AI generated ...6.4 Notch Signaling In... · 6.4. 1 Intestinal Phenotypes... · 6.4. 2.1 Atoh1 Promotes...
  10. [10]
    The Nobel Prize in Physiology or Medicine 1971 - Press release
    Cyclic AMP was discovered as the “second messenger” in relation to hormonal effects. It therefore came as no surprise when Sutherland in 1965 found that the ...
  11. [11]
    The Evolution of Quorum Sensing in Bacterial Biofilms | PLOS Biology
    Quorum sensing is also phylogenetically widespread, which suggests an early origin in bacterial evolution [19].
  12. [12]
    The Evolutionary History of Quorum-Sensing Systems in Bacteria
    The most likely timing for this origin is after the divergence of Xanthomonas species and Xylella, which lack homologs (Bhattacharyya et al. 2002), but before ...
  13. [13]
    The Evolution of Two-Component Systems in Bacteria Reveals ...
    Two-component systems including histidine protein kinases represent the primary signal transduction paradigm in prokaryotic organisms.
  14. [14]
    Conservation and Reiteration of a Kinase Cascade - PubMed
    A cascade of three protein kinases has emerged as a conserved functional module in a wide variety of signal transduction pathways in diverse organisms.
  15. [15]
    Diversity in Chemotaxis Mechanisms among the Bacteria and Archaea
    In bacteria and archaea, motility is controlled by a two-component system involving a histidine kinase that senses the environment and a response regulator, a ...
  16. [16]
    Plants: the latest model system for G‐protein research | EMBO reports
    Genes that encode G‐protein‐signalling elements have been identified in slime moulds, fungi, plants and animals. Several recent reviews have focused on the ...<|separator|>
  17. [17]
    Comparative analysis of the repertoire of G protein-coupled ...
    May 16, 2013 · Members of class VI of fungal GPCRs are characterized by the presence of both 7-transmembrane regions and an RGS (regulator of G protein ...
  18. [18]
    Evolution of Two-Component Signal Transduction - Oxford Academic
    TCST systems are of bacterial origin and radiated into archaea and eukaryotes by lateral gene transfer. Their components show extensive coevolution.
  19. [19]
    The Evolution of Two-Component Signal Transduction Systems - PMC
    The evolution of two-component systems in bacteria reveals different strategies for niche adaptation. PLoS Comput Biol. 2006;2:e143. doi: 10.1371/journal ...
  20. [20]
    Pathways of Intracellular Signal Transduction - The Cell - NCBI - NIH
    Figure 13.18. Synthesis and degradation of cAMP. Cyclic AMP is synthesized from ATP by adenylyl cyclase and degraded to AMP by cAMP phosphodiesterase. How does ...
  21. [21]
    Phosphoinositide-specific phospholipase C in health and disease
    PIP2, which is located within the plasma membrane, is cleaved by PI-PLC enzymes, generating the two well-known second messengers, DAG and IP3. DAG remains bound ...
  22. [22]
    Fundamentals of Cellular Calcium Signaling: A Primer - PMC
    Ionized calcium (Ca2+) is the most versatile cellular messenger. All cells use Ca2+ signals to regulate their activities in response to extrinsic and intrinsic ...
  23. [23]
    Second Messenger System - an overview | ScienceDirect Topics
    Second messenger systems, including Ca2+ and cAMP, are essential for transforming synaptic stimulation into long-lasting neuronal representations and memory- ...Major Second Messengers... · Signal Transduction Pathways...
  24. [24]
    The Cyclic AMP Pathway - PMC - NIH
    cAMP generated as a consequence of AC activation can activate several effectors, the most well studied of which is cAMP-dependent protein kinase (PKA) (Pierce ...
  25. [25]
    Intercellular Ca2+ Waves: Mechanisms and Function - PMC
    Intercellular calcium (Ca 2+ ) waves (ICWs) represent the propagation of increases in intracellular Ca 2+ through a syncytium of cells.
  26. [26]
    Principles of Chemical Signaling and Communication by Microbes
    Autocrine signaling occurs when a ligand acts on the same cell that releases it. Paracrine signaling occurs when a ligand diffuses in a small area and only acts ...
  27. [27]
    Physiology, Cellular Messengers - StatPearls - NCBI Bookshelf
    Apr 24, 2023 · Introduction. Cellular communication is a complex ... The extracellular calcium-sensing receptor and cell-cell signaling in epithelia.
  28. [28]
    Paracrine Factors - Developmental Biology - NCBI Bookshelf
    When proteins synthesized by one cell can diffuse over small distances to induce changes in neighboring cells, the event is called a paracrine interaction, and ...
  29. [29]
    Autocrine TGF-β in Cancer: Review of the Literature and Caveats in ...
    Jan 19, 2021 · This review surveys the many observations of autocrine TGF-β signaling in tumor biology, including data from cell culture and animal models as well as from ...
  30. [30]
    An autocrine ActivinB mechanism drives TGFβ/Activin signaling in ...
    Jul 22, 2019 · In many different cancers, TGFβ pathway activation involves autocrine loops, due to the high expression of genes encoding the different TGFβ ...
  31. [31]
    Wound repair and regeneration: Mechanisms, signaling ... - Science
    Another growth factor, PDGF, has proven effective in stimulating healing in patients. In 1997, becaplermin, a recombinant human PDGF (rhPDGF-BB), was approved ...Wound Repair And... · Wound-Healing Pathology · Current Treatments And Their...
  32. [32]
    Paracrine IL-2 is required for optimal type 2 effector cytokine ...
    IL-2 is a pleiotropic cytokine that promotes the differentiation of T helper (Th) cell subsets including Th1, Th2, and Th9 cells, but impairs development of Th ...
  33. [33]
    Paracrine communication maximizes cellular response fidelity in ...
    Oct 8, 2015 · Cells that are damaged during wounding immediately release a molecule called ATP, which acts as a danger signal to activate the wound healing ...Missing: PDGF | Show results with:PDGF
  34. [34]
    Physiology, Stress Reaction - StatPearls - NCBI Bookshelf
    May 7, 2024 · A stress response is mediated through a complex interplay of nervous, endocrine, and immune mechanisms, activating the sympathetic-adreno-medullar (SAM) axis.
  35. [35]
    Physiology, Thyroid Hormone - StatPearls - NCBI Bookshelf
    Jun 5, 2023 · In general, when the thyroid hormone binds to its intranuclear receptor, it activates the genes for increasing metabolic rate and thermogenesis.
  36. [36]
    Juxtacrine Signaling - Developmental Biology - NCBI Bookshelf
    A ligand (Delta, Jagged, or Serrate protein) on one cell binds to the extracellular domain of the Notch protein on an adjacent cell. This binding causes a shape ...
  37. [37]
    Juxtacrine Signaling Is Inherently Noisy - PMC - NIH
    (A) An illustration of axon guidance mediated by ephrin signaling. During axon development, axons (yellow) that express Eph receptors in their growth cones ...
  38. [38]
    Principles of endocrinology - NCBI - NIH
    Classification of hormones and chemical signalling mechanisms ... distant target tissue defines the 'classical' endocrine and neuroendocrine signalling mechanism.Missing: bloodstream | Show results with:bloodstream
  39. [39]
    Hormones, Receptors and Target Cells
    Endocrine action: the hormone is distributed in blood and binds to distant target cells. Paracrine action: the hormone acts locally by diffusing from its ...Missing: bloodstream | Show results with:bloodstream
  40. [40]
    Nuclear receptors in health and disease: signaling pathways ...
    Jul 28, 2025 · Dysregulation of nuclear receptor signaling is implicated in the pathogenesis of numerous diseases, including cancers, metabolic disorders, ...
  41. [41]
    Nuclear receptors and inflammation control: molecular mechanisms ...
    Nuclear receptors can interfere with numerous mechanisms required for signal-dependent gene activation so as to suppress inflammatory responses. Representative ...
  42. [42]
    Retinoid X receptor heterodimers in hepatic function - Frontiers
    RXR is widely involved in hepatic glucose metabolism, lipid metabolism, cholesterol metabolism and bile acid homeostasis as well as hepatic fibrosis.
  43. [43]
    Function and Evolution of Nuclear Receptors in Environmental ...
    During evolution, these multipurpose lipid sensors presumably lost the ligand-based regulation of transcriptional activation secondarily by duplications and ...
  44. [44]
    The Molecular Basis of G Protein–Coupled Receptor Activation - PMC
    GPCR activation involves a ligand binding, promoting GTP exchange, leading to G protein dissociation and downstream signals. Agonist binding stabilizes the ...Missing: seminal | Show results with:seminal
  45. [45]
    Ultrasensitivity in Phosphorylation-Dephosphorylation Cycles ... - NIH
    Aug 8, 2013 · Sequential cascades of phosphorylation cycles, such as cascades of MAP kinase, can amplify signals and increase the degree of ...
  46. [46]
    Complex roles of cAMP–PKA–CREB signaling in cancer
    Nov 24, 2020 · cAMP–PKA signaling can regulate cancer cell growth, migration, invasion and metabolism. This review highlights the important roles of cAMP–PKA–CREB signaling ...
  47. [47]
    MAPK signal pathways in the regulation of cell proliferation ... - Nature
    Mar 1, 2002 · Mitogen-activated protein kinase (MAPK) cascades have been shown to play a key role in transduction extracellular signals to cellular responses.
  48. [48]
    Targeting Aberrant RAS/RAF/MEK/ERK Signaling for Cancer Therapy
    The RAS/RAF/MEK/ERK (MAPK) signaling cascade is essential for cell inter- and intra-cellular communication, which regulates fundamental cell functions such as ...
  49. [49]
    Targeting PI3K/Akt signal transduction for cancer therapy - Nature
    Dec 16, 2021 · As a lipid phosphatase, PTEN negatively regulates the PI3K signaling pathway and transforms PIP3 into PIP2. When PTEN is mutated or participates ...
  50. [50]
    PTEN Tumor Suppressor Network in PI3K-Akt Pathway Control - PMC
    This review will highlight the networking of PTEN with other inhibitors of the pathway, relevant to cancer progression.
  51. [51]
    Notch signaling pathway: architecture, disease, and therapeutics
    NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription ...Missing: post- | Show results with:post-
  52. [52]
    The Notch signaling network in muscle stem cells during ...
    Apr 22, 2022 · In this review, we discuss the diverse ways by which Notch signaling factors and other modifying partners are operating during the lifetime of muscle stem ...
  53. [53]
    Wnt/β-catenin signalling: function, biological mechanisms ... - Nature
    Jan 3, 2022 · The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis.
  54. [54]
    TGF-β Signaling from Receptors to Smads - PMC - PubMed Central
    The TGF-β family members signal via heteromeric complexes of type I and type II receptors, which activate members of the Smad family of signal transducers.
  55. [55]
    Cross-talk between mitogenic Ras/MAPK and survival PI3K/Akt ...
    Jan 19, 2012 · In the present paper, we describe multiple levels of cross-talk between the PI3K (phosphoinositide 3-kinase)/Akt and Ras/MAPK (mitogen-activated protein kinase ...Introduction · Cross-talk between Ras/MAPK... · Context-dependent cross-talk
  56. [56]
    Computational Modeling of PI3K/AKT and MAPK Signaling ...
    There are multiple cross-talk points between these two pathways, whose coordinated action determines the cell fate [11]. It is not surprising that the PI3K/AKT ...
  57. [57]
    Glutamate Receptor Ion Channels: Structure, Regulation, and Function
    Glutamate receptors mediate fast excitatory synaptic transmission in the central nervous system and are localized on neuronal and non-neuronal cells. These ...
  58. [58]
    Rho GTPases: Central regulators of cell migration - PMC
    Rho family GTPases regulate the cytoskeletal rearrangements and adhesions necessary for cell shape change and cellular adhesion.
  59. [59]
    Insulin Signaling and the Regulation of Glucose Transport
    Oct 30, 2005 · Insulin stimulates the translocation of a pool of Glut4 to the plasma membrane, through a process of targeted exocytosis. The microtubule ...
  60. [60]
    Calcium-Induced Calcium Release in Skeletal Muscle
    CICR is defined as Ca 2+ release by the action of Ca 2+ alone without the simultaneous action of other activating processes.
  61. [61]
    Cell Biology of Ca2+-Triggered Exocytosis - PMC - PubMed Central
    Ca2+ triggers many forms of exocytosis in different types of eukaryotic cells, for example synaptic vesicle exocytosis in neurons, granule exocytosis in mast ...
  62. [62]
    Seizures and Epilepsies due to Channelopathies ... - PubMed Central
    Certain central nervous system proteins have been involved in epilepsy or acute neurological diseases with seizures either due to underlying gene defects or ...
  63. [63]
    The JAK/STAT signaling pathway: from bench to clinic - Nature
    Nov 26, 2021 · JAKs are noncovalently associated with cytokine receptors, mediate tyrosine phosphorylation of receptors, and recruit one or more STAT proteins.
  64. [64]
    NF-κB in biology and targeted therapy: new insights and ... - Nature
    Mar 4, 2024 · IκB is phosphorylated by the active IKK complex, which causes ubiquitination and eventual destruction of IκB. NF-κB dimer is released and ...
  65. [65]
    Structural and functional relationships of the steroid hormone ...
    Steroid hormone receptors are members of a family of ligand inducible transcription factors, and regulate the transcriptional activation of target genes by ...Introduction · Steroid Receptors Contain... · Steroid Receptors' Af1...
  66. [66]
    revealing the importance of the IFN JAK/STAT pathway - PMC - NIH
    This signalling pathway results in the expression of hundreds of effector genes known as interferon stimulated genes (ISGs). These genes are the basis for an ...
  67. [67]
    Collaboration between distinct SWI/SNF chromatin remodeling ...
    Aug 13, 2024 · Our studies demonstrate that SWI/SNF complexes regulate enhancer selection and activation in response to inflammatory signals. Disruption of ...
  68. [68]
    Article The Transcriptional Coactivators p300 and CBP Are Histone ...
    p300/CBP is a transcriptional adaptor that integrates signals from many sequence-specific activators via direct interactions.
  69. [69]
    Epigenetic mechanisms to propagate histone acetylation by p300/CBP
    Jul 17, 2023 · We propose a model in which p300/CBP replicates histone N-terminal tail acetylation within the H3-H4 tetramer to inherit epigenetic storage.
  70. [70]
    The Notch intracellular domain integrates signals from Wnt ...
    Notch signaling is a highly conserved signal transduction pathway that regulates stem cell maintenance and differentiation in several organ systems.
  71. [71]
    Pathway centric analysis for single-cell RNA-seq and spatial ...
    Dec 18, 2023 · Using pathway gene sets, we show that GSDensity can accurately detect biologically distinct cells and reveal novel cell-pathway associations ...
  72. [72]
    GPCR Desensitization: Acute and Prolonged Phases - PMC
    Short-term desensitization occurs over minutes, and is primarily associated with β-arrestins preventing G protein interaction with a GPCR.Missing: paper | Show results with:paper
  73. [73]
    GPCR Signaling Regulation: The Role of GRKs and Arrestins
    Here we focus on the mechanisms of GRK- and arrestin-mediated regulation of GPCR signaling, which includes homologous desensitization and redirection of ...
  74. [74]
    Endocytosis of Receptor Tyrosine Kinases - PMC - NIH
    Signaling from receptor tyrosine kinases (RTKs) is regulated by endocytosis. Ligand binding at the cell surface triggers internalization and subsequent sorting ...Missing: seminal paper
  75. [75]
    Opioid receptor desensitization: mechanisms and its link to tolerance
    This review will summarize receptor-related mechanisms that could underlie tolerance especially receptor desensitization.
  76. [76]
    EGFR Downregulation after Anti-EGFR Therapy Predicts the ...
    We also demonstrated that downregulation of EGFR on the cell surface after anti-EGFR antibody treatment is caused by augmented degradation of antibody-bound ...
  77. [77]
    Biased agonism: An emerging paradigm in GPCR drug discovery
    Biased agonism, or functional selectivity, is when ligands selectively activate one pathway of a GPCR over another, changing the linear spectrum of GPCR ...Missing: post- | Show results with:post-
  78. [78]
    Computational systems biology in disease modeling and ... - Nature
    Oct 3, 2022 · This review introduces biomedical applications and analyses of interactions among gene, protein and drug molecules for modeling disease mechanisms and drug ...Network Structure In Systems... · Graph-Based Learning For... · Analysis Of Dynamic Modeling
  79. [79]
    Positive and negative regulation of insulin signaling through IRS-1 ...
    These agents negatively regulate the IRS1 functions by phosphorylation but also via others molecular mechanisms (SOCS expression, IRS degradation, O-linked ...
  80. [80]
    Protein phosphatase 2A, a negative regulator of the ERK signaling ...
    Protein phosphatase 2A, a negative regulator of the ERK signaling pathway, is activated by tyrosine phosphorylation of putative HLA class II-associated protein ...Missing: cascades feedback
  81. [81]
    Relaxation oscillations and hierarchy of feedbacks in MAPK signaling
    Jan 3, 2017 · A positive feedback loop involving RAS and SOS, which leads to bistability and allows for switch-like responses to inputs, is nested within a ...
  82. [82]
    Cross-talk of WNT and FGF signaling pathways at GSK3beta to ...
    ... PI3K-AKT signaling cascade to downregulate GSK3beta activity depending on Ser 9 phosphorylation. Because GSK3beta-dependent phosphorylation of beta-catenin ...
  83. [83]
    Signalling scaffolds and local organization of cellular behaviour - Nature Reviews Molecular Cell Biology
    ### Summary: Scaffold Proteins in Signaling Networks and Feedback Mechanisms
  84. [84]
    Rhythmic PER abundance defines a critical nodal point for negative ...
    Nov 13, 2009 · Circadian rhythms in mammals are generated by a transcriptional negative feedback loop that is driven primarily by oscillations of PER and CRY, ...
  85. [85]
    Treg and CTLA-4: two intertwining pathways to immune tolerance
    Jul 10, 2013 · In this article, I suggest that CTLA-4 and Treg represent complementary and largely overlapping mechanisms of immune tolerance.Missing: feedback | Show results with:feedback
  86. [86]
    Interleukin-17 promotes autoimmunity by triggering a ... - PubMed
    Oct 17, 2008 · Dysregulated cytokine expression and signaling are major contributors to a number of autoimmune diseases ... feedback loop via interleukin ...