Fact-checked by Grok 2 weeks ago

Intracrine

Intracrine signaling is a mode of cellular communication in which hormones, growth factors, or other signaling molecules exert their effects intracellularly within the same where they are synthesized or after uptake from neighboring cells, bypassing traditional cell surface receptors to directly interact with cytosolic or targets. This contrasts with endocrine signaling (distant targets via bloodstream), (adjacent cells), and classical (via surface receptors on the producing ), enabling rapid, localized regulation of processes like and protein synthesis without extracellular release. The term "intracrine" was coined in to describe such internal actions, particularly of peptide hormones like angiotensin II. The concept emerged from studies on intracellular trafficking of signaling molecules, initially focusing on angiotensin II's nuclear accumulation in vascular smooth muscle cells, which suggested feed-forward regulatory loops independent of plasma membrane interactions. Over the subsequent decades, research has expanded intracrinology to encompass a diverse array of molecules, including not only peptides but also steroid hormones such as estrogens and androgens, which are locally synthesized from precursors like or dehydroepiandrosterone (DHEA) via enzymes including and . These intracrines often translocate to the or , modulating transcription factors and cellular , while mechanisms like exosome-mediated transfer or cytoskeletal nanotubes facilitate intercellular spread without . Key examples of intracrine molecules include angiotensin II, which regulates cardiovascular and through intracellular renin-angiotensin system activation; vascular endothelial growth factor (VEGF), promoting cell survival and differentiation in and maintenance without inducing ; and parathyroid hormone-related protein (PTHrP), influencing and cancer via nuclear signaling. Sex steroids exemplify intracrine actions in non-gonadal tissues, where immune cells like macrophages synthesize and respond to estrogens to fine-tune , production, and , impacting conditions such as autoimmune diseases and post-traumatic responses. Additionally, fibroblast growth factor 2 (FGF2) and angiogenin drive developmental processes, including cardiac embryogenesis and tissue repair. Physiologically, intracrine signaling plays critical roles in , , and , such as enhancing insulin sensitivity in via local estrogen production or promoting tumor through PTHrP's localization signal. Dysregulation contributes to diseases including cancer, where intracrine VEGF sustains tumor cell survival, and metabolic disorders, underscoring its therapeutic potential in targeted interventions that modulate intracellular pathways.

Definition and Concepts

Definition of Intracrine Signaling

Intracrine signaling refers to the mechanism by which signaling molecules, such as peptides, growth factors, or hormones, are synthesized within a and exert their biological effects directly inside that same , without being secreted into the . These ligands bind to intracellular receptors located in the or , thereby influencing processes like transcription, enzymatic activation, or other intracellular pathways. This mode of action allows for precise, compartment-specific that bypasses the need for extracellular diffusion or . A defining characteristic of intracrine signaling is the absence of ligand secretion, which contrasts with traditional hormone signaling paradigms that rely on release and subsequent to surface receptors. Instead, intracrine s may be retained in the producer through mechanisms such as alternative translation from atypical start sites, preventing incorporation into the secretory pathway, or by acting after limited . Representative examples include the intracellular retention of fibroblast growth factors (FGFs), which can translocate to the to modulate , and the local conversion of precursors like dehydroepiandrosterone (DHEA) into active s within target tissues. These features enable intracrine signaling to function independently of circulating levels, providing a layer of in cellular responsiveness. The biological significance of intracrine signaling lies in its capacity for rapid, localized control over key cellular functions, including , , , and metabolic . By operating within confined intracellular compartments, it facilitates immediate responses to endogenous cues, such as or developmental signals, without the delays associated with intercellular communication. This is particularly evident in tissues with high metabolic demands, where intracrine pathways fine-tune and prevent over-reliance on distant endocrine sources. Unlike , which requires secretion and re-uptake by the same , intracrine action remains fully intracellular, enhancing efficiency in dynamic environments. The concept of intracrine signaling was first introduced in 1984 by Richard N. Re and his laboratory to describe the intracellular action of hormones, as opposed to their effects via surface receptors. This term was later broadened in the through Labrie's work on intracrinology, emphasizing the role of local steroidogenesis in peripheral tissues for hormone production and action.

Distinction from Other Signaling Types

Intracrine signaling fundamentally differs from endocrine signaling, where hormones such as insulin are secreted into the bloodstream to act on distant target s via surface receptors. In contrast, intracrine actions occur entirely within the , bypassing circulation and external secretion to enable direct intracellular regulation without systemic dissemination. Unlike , which involves the local diffusion of ligands like to influence nearby cells during processes such as , intracrine signaling is confined to the interior of a single , avoiding intercellular spread and focusing on internal compartmentalized responses. This strict intracellular localization distinguishes it from paracrine mechanisms that rely on short-range extracellular gradients. Autocrine signaling, exemplified by platelet-derived growth factor (PDGF) binding back to surface receptors on the same cell after secretion, contrasts with intracrine signaling in that the latter typically involves non-secreted ligands acting directly on intracellular targets, such as nuclear receptors, without an extracellular phase. While both can affect the producing cell, intracrine pathways emphasize internal action independent of secretion and re-uptake. Juxtacrine signaling requires direct physical contact between cells, as seen in pathway activation through membrane-bound ligands, whereas intracrine signaling operates without cell-cell interaction, relying solely on endogenous or internalized molecules within the cell's interior. This non-contact nature underscores intracrine's autonomy from extracellular cues. Although overlaps exist in hybrid forms, such as internalized ligands enabling effects after initial , pure intracrine signaling prioritizes non-secretory, intracellular mechanisms to maintain precise . The evolutionary advantage of intracrine signaling lies in its ability to provide compartmentalized, feedback-independent regulation, particularly beneficial in constrained microenvironments like tumors—where (VEGF) promotes cell survival internally—or during tissue development, allowing sustained, autonomous responses without reliance on external signals.

Historical Development

Origin and Evolution of the Term

The term "intracrine" was first introduced in 1984 by Richard N. Re and colleagues to describe the intracellular action of peptide hormones within their cell of synthesis, contrasting with traditional extracellular signaling at cell surface receptors. This concept emerged from observations in cardiovascular tissues, where hormones like II were found to exert effects intracellularly, challenging the dominant endocrine and paracrine paradigms. The initial published elaboration appeared in 1989, highlighting intracrine mechanisms in the cellular biology of and their role in cardiovascular regulation. In the 1990s, the term expanded significantly through the work of Fernand Labrie and his team, who applied it to local steroidogenesis in peripheral tissues, emphasizing tissue-specific synthesis and action of hormones such as androgens and estrogens without reliance on circulating levels. This development led to the establishment of "intracrinology" as a distinct field focused on the intracellular formation and inactivation of steroids to achieve precise local control. A seminal publication was Labrie's 1991 review in Molecular and Cellular Endocrinology, which detailed intracrine androgen production in the prostate and advocated shifting research from systemic circulating hormones to local intracrine processes. Labrie's reviews in the 1990s further reinforced this evolution, underscoring how intracrine mechanisms explained variations in hormone effects across tissues that classical endocrinology could not adequately account for. By the , intracrine signaling had integrated into broader and literature, with Re's 2002 paper proposing a theoretical framework linking it to ancient regulatory mechanisms for cellular processes like synthesis. The marked wider recognition in fields like cancer and cardiovascular research, as evidenced by Labrie's 2015 retrospective on three decades of intracrinology, which highlighted its implications for tissue-specific therapies. This progression was driven by the need to address limitations in classical , particularly its inability to explain diverse, localized responses in and .

Shift to Intracrinology

The transition to intracrinology marked a significant in , moving away from the classical model that emphasized s circulating in the bloodstream to act on distant target cells via extracellular receptors. Instead, intracrinology focuses on the local synthesis, action, and inactivation of s within the same peripheral target cells, utilizing precursors like dehydroepiandrosterone (DHEA) to generate active steroids such as androgens and estrogens without substantial release into circulation. This framework reduces dependence on systemic levels and highlights tissue-specific regulation, allowing cells to tailor activity to local needs while minimizing off-target effects. A key proponent of this shift was Fernand Labrie, whose work in the strongly advocated for intracrine steroidogenesis, particularly in the contexts of aging and disease. Labrie emphasized that in postmenopausal women, nearly 100% of active sex steroids are produced intracrinely in peripheral tissues from adrenal precursors, addressing deficiencies in gonadal production. His research integrated intracrinology with emerging genomic insights, demonstrating cell-specific expression of steroidogenic enzymes; for instance, (CYP19A1) is highly expressed in breast tissue, enabling local biosynthesis that influences tissue physiology and pathology. This genomic perspective revealed polymorphisms in genes like CYP19A1 associated with altered metabolism and increased risk, underscoring the field's molecular foundation. The advent of intracrinology profoundly impacted research directions, spurring investigations into therapeutic applications of precursors like (DHEA). Clinical studies have shown that intravaginal prasterone administration leads to local conversion into active steroids, improving vaginal atrophy in postmenopausal women without elevating systemic hormone levels, thus validating the intracrine paradigm's clinical relevance. Reviews from the 2010s, including reflections on the field's three-decade , further formalized intracrinology by synthesizing evidence from cloning and peripheral analyses. It also overcame longstanding challenges, such as discrepancies between low androgen concentrations and robust tissue responses; in , for example, local dihydrotestosterone (DHT) levels persist at activating concentrations despite castrate androgens, attributable to intracrine biosynthesis from adrenal sources. As of 2025, intracrinology is established as a recognized subdiscipline within , with active research integrated into broader studies and featured in major journals and conferences. Ongoing work explores its implications for , such as targeted precursor therapies, while annual events like the Endocrine Society's meeting continue to advance discussions on dynamics.

Molecular Mechanisms

Intracellular Localization and Receptors

Intracrine ligands achieve intracellular localization through specific mechanisms that enable them to remain within the producing cell or target intracellular compartments, bypassing extracellular secretion. Many intracrine ligands, such as fibroblast growth factors (FGFs), possess nuclear localization signals (NLS) that facilitate into the via importin-mediated pathways. For instance, FGF1 contains a functional NLS sequence (KKPK, 23-27) essential for its nuclear translocation and subsequent intracrine activities in neuronal cells. Similarly, FGF2 and FGF3 incorporate NLS motifs that direct them to the , supporting roles in and independent of extracellular signaling. Cytoplasmic retention of these ligands often occurs through interactions with binding proteins that sequester them away from secretory pathways, as observed with intracellular partners of FGF1 and FGF2 that modulate their availability for nuclear entry. Organelle-specific targeting, such as to mitochondria, is prominent in steroid hormone precursors, where locally synthesized intermediates like dehydroepiandrosterone (DHEA) are converted to within the same compartment to exert regulatory effects. Intracellular receptors for intracrine ligands encompass both and non-nuclear types, enabling diverse signaling modalities within the cell. receptors, particularly those for steroid hormones such as the (AR) and (ER), bind lipophilic intracrine ligands like testosterone and directly in the or , initiating . These receptors undergo ligand-induced conformational changes that promote dimerization and recruitment of co-activators to enhance . Non-nuclear receptors include cytoplasmic s activated by internalized peptide ligands; for example, angiotensin II can stimulate intracellular pools of its receptors to phosphorylate cytoplasmic targets via kinase cascades. In the case of (PTHrP), nuclear variants of the PTH1 receptor (PTHR1) or direct ligand-receptor interactions in the mediate proliferative effects in vascular cells, distinct from surface receptor functions. Binding of intracrine ligands to their intracellular receptors typically induces conformational shifts that facilitate co-activator or co-repressor , leading to altered transcription or protein modifications, without reliance on G-protein coupling characteristic of plasma membrane receptors. For nuclear receptors like and , ligand binding exposes interaction surfaces for transcriptional machinery, amplifying intracrine signals in contexts like tissue-specific hormone regulation. This activation contrasts with extracellular signaling by allowing rapid, localized responses to on-site ligand production. Evidence for intracrine localization and receptor interactions derives from multiple experimental approaches demonstrating co-localization and functional specificity. studies have revealed nuclear accumulation of PTHrP in vascular cells, with a diffuse reticulated pattern in approximately 5-6% of nuclei in overexpressing cells, confirming NLS-dependent targeting. Co-localization of FGF1 with markers in PC12 cells via and imaging supports its intracrine role in anti-apoptotic signaling. and mutant models further delineate these effects; deletion of the PTHrP NLS abolishes targeting and reduces by 4-5 fold, while PTHrP-null embryos exhibit impaired vascular without alterations in secretory pathways. Similarly, FGF1 mutants lacking the NLS fail to exert neurotrophic effects intracellularly, despite intact receptor capability. Due to on-site , intracellular ligand concentrations in intracrine systems often significantly exceed circulating serum levels, enabling potent local effects that are not reflected in systemic measurements.

Signal Transduction Pathways

Upon to intracellular receptors, intracrine ligands initiate diverse cascades that propagate within the , distinct from classical paracrine or autocrine mechanisms due to their confinement to the intracellular compartment. These pathways often involve receptor kinases (RTKs) or other intracellular binding sites, leading to events that activate downstream effectors. For instance, intracrine 1 (FGF1), which lacks a secretory signal and contains a , promotes anti- and neurotrophic effects through nuclear translocation and interactions with intracellular partners like ribosomal proteins and Rheb, independent of FGFR activation, in neuronal cells. Similarly, the PI3K/Akt pathway is engaged by intracrine (VEGF), particularly via VEGFR1 or VEGFR2, to enhance survival by inhibiting in cancer and stem cells, as evidenced by reduced Akt upon VEGF knockdown. Nuclear effects of intracrine signaling prominently involve direct modulation of transcription factors. Intracrine VEGF-VEGFR complexes translocate to the in endothelial and tumor cells, where they may modulate transcription factors and involved in cell and adaptation. This nuclear localization has been observed in various cell types, including osteoblasts and cells, supporting roles in and . Epigenetic modifications, such as histone or , may also be influenced by these intracrine signals, altering accessibility for sustained changes, though specific mechanisms remain under investigation in contexts like intracrine actions. Recent studies (as of 2025) have identified additional intracrine roles, such as VEGF signaling in maintaining adult hippocampal and , and GPCR-mediated signaling at intracellular sites like droplets. Non-genomic actions occur rapidly in the , bypassing transcriptional changes. For example, intracrine angiotensin II stimulates inward calcium fluxes through activation and modulation of voltage-dependent calcium channels in myocytes, enabling quick responses like or . modulation by intracrine ligands, such as VEGF affecting mitochondrial calcium handling, further supports metabolic regulation and prevention of . Cross-talk between intracrine-initiated pathways and others amplifies signaling integration. Intracrine VEGF depletion activates tyrosine phosphatases that inhibit and c-MET, linking to reduced MAPK/ERK activity in cells. Intracrine signaling, activated by ligands like FGF, intersects with Wnt/β-catenin by modulating β-catenin stability through ERK-mediated , influencing fate decisions. Experimental validation of these pathways relies on targeted perturbations and techniques. siRNA knockdown of intracrine VEGF specifically inhibits MAPK/ERK and PI3K/Akt in colorectal and cells, confirming pathway dependence without affecting extracellular signaling. Live-cell with fluorescently labeled VEGF165 has tracked its intracellular trafficking and nuclear accumulation, revealing real-time signal propagation in endothelial cells.

Positive Feedback Loops

In intracrine signaling, loops arise when a acts intracellularly to induce its own synthesis or upregulate receptor expression, thereby amplifying the signal without requiring external stimuli. This self-reinforcing mechanism enhances cellular responsiveness and can establish persistent states by creating feed-forward cycles within the cell. For instance, intracellular angiotensin II (Ang II) binds to nuclear angiotensin type 1 receptors, promoting transcription of renin and angiotensinogen genes, which in turn increases endogenous Ang II production and local (ACE) expression in renal tubular cells via AT1 receptor activation. Representative examples illustrate these loops across systems. In steroid intracrinology, dehydroepiandrosterone (DHEA) is converted intracellularly to active androgens or estrogens by enzymes such as and , with the resulting steroids capable of upregulating these biosynthetic enzymes in peripheral tissues like breast epithelium, sustaining action. Similarly, (VEGF) forms an intracrine loop under hypoxic conditions, where intracellular VEGF binds VEGFR2 to prevent and promote nuclear accumulation, further enhancing VEGF expression and supporting in endothelial cells. Mathematical models of these loops often capture their amplifying nature through differential equations representing -receptor interactions. A model for ligand concentration [L] dynamics is given by: \frac{d[L]}{dt} = k [R] [L] - \delta [L] where k is the rate constant for ligand-induced or receptor , [R] is receptor concentration, and \delta is the degradation rate. occurs when k [R] > \delta, leading to in [L] until saturation or inhibition; steady-state analysis yields [L] = 0 (quiescent) or unbounded growth (unstable without regulators), with inhibitors stabilizing low states. This framework, adapted from autocrine models, highlights and context-dependent signaling in intracrine systems. Biologically, these loops sustain chronic physiological states, such as via persistent Ang II signaling, while dysregulation contributes to pathologies like therapeutic in cancer, where VEGF loops enable tumor under . from time-course studies demonstrates signal rises; for example, in EGFR autocrine systems akin to intracrine feedback, positive loops increase ligand release rates fourfold over hours, prolonging MAPK activation. Inhibitors disrupt these cycles, as ACE inhibitors block Ang II-mediated RAS upregulation, reducing intrarenal feedback and alleviating .

Intracrine in

Angiotensin II Actions

Angiotensin II (Ang II) exerts intracrine actions within cardiac and vascular cells, where it is synthesized intracellularly through local components of the renin-angiotensin system (RAS), including renin and (), independent of circulating precursors. In human cardiac tissue, particularly the left ventricle and atria, Ang II production occurs via noncanonical pathways, such as chymase-mediated processing of angiotensin-(1-12), leading to elevated intracellular levels in diseased states like and . These intracellular Ang II molecules interact with nuclear angiotensin type 1 (AT1) and type 2 (AT2) receptors localized on the inner and within the nucleoplasm, facilitating direct modulation of without requiring extracellular receptor engagement. Intracrine Ang II activates key signal transduction pathways in cardiac fibroblasts and cardiomyocytes, distinct from its classical extracellular effects. In fibroblasts, nuclear AT1 receptors trigger mitogen-activated protein kinase (MAPK) signaling through inositol trisphosphate receptor (IP3R)-mediated calcium mobilization, promoting hypertrophic responses, while AT2 receptors stimulate nuclear factor-kappa B (NF-κB) via nitric oxide production to regulate inflammatory and fibrotic gene transcription. In cardiomyocytes, intracrine Ang II influences ion channel activity, enhancing intracellular calcium influx via sarcolemmal channels and sarcoplasmic reticulum release, which contributes to cellular excitability and contractile remodeling. These pathways underscore the role of intracrine Ang II in maladaptive cardiac responses, such as those observed in pressure overload. The cellular effects of intracrine Ang II prominently include promotion of proliferation and synthesis, leading to interstitial , as evidenced by upregulated collagen-1A1 mRNA expression and increased secretion in atrial from models. In cardiomyocytes, it induces structural remodeling, including and altered intercellular communication via gap junctions, exacerbating ventricular dysfunction. Studies from the early 2020s, such as those examining NADPH oxidase-mediated in cardiac , confirm these effects contribute to pathological in hypertensive hearts. Transgenic models overexpressing cardiac components demonstrate that targeted intracellular blockade, such as with cell-impermeant AT1 inhibitors, significantly reduces and , highlighting the intracrine pathway's contribution beyond extracellular signaling. A critical aspect of intracrine Ang II is its independence from levels, allowing sustained signaling in cardiac tissue even when circulating is suppressed, which explains partial resistance to receptor blockers (ARBs) in certain hypertrophic conditions. For instance, in models, intracellular Ang II persists despite extracellular AT1 blockade with agents like , maintaining fibrotic progression through activation. This autonomy of local intracrine underscores its role in ARB-refractory cardiac remodeling. (PTHrP) is synthesized and retained within cardiomyocytes, where it exerts intracrine effects by binding to a of its type 1 receptor (PTH1R). This local production allows PTHrP to act autonomously within the cell, independent of extracellular secretion, facilitating rapid responses to mechanical or ischemic stress in the myocardium. The localization of PTHrP and PTH1R enables direct modulation of and cellular processes, distinguishing it from classical secretory pathways. In its intracrine mode, PTHrP promotes cardiomyocyte survival through cAMP-independent signaling pathways, such as activation of (PKC) and casein kinase 2 (CK2), which inhibit mitochondrial-dependent . During ischemia, PTHrP suppresses by attenuating and preserving cellular integrity, thereby enhancing resistance to ischemia-reperfusion injury in myocardial cells. PTHrP plays a critical role in myocardial adaptation to by enhancing contractility in post-ischemic and promoting vascular relaxation in coronary vessels, which supports overall cardiac performance. These effects contribute to adaptive responses, including those during physiological states like , where PTHrP-mediated aids in accommodating increased and blood volume. Evidence from models underscores PTHrP's importance; mice lacking the PTH/PTHrP receptor exhibit impaired myocardial growth and abrupt cardiomyocyte death during development, highlighting its essential role in cardiac and survival. While PTHrP can function in both paracrine and intracrine manners, its intracrine actions predominate in autoprotective mechanisms within cardiomyocytes, enabling localized regulation of growth and stress responses without reliance on intercellular signaling.

Vascular Endothelial Growth Factor (VEGF)

(VEGF), particularly VEGF-A and VEGF-B isoforms, is autoproduced within endothelial cells and cardiomyocytes, where it exerts intracrine effects by binding to cytoplasmic VEGF receptors (VEGFRs), such as VEGFR-2, independent of extracellular secretion. This intracellular signaling sustains cellular and vascular integrity in the heart, contrasting with paracrine VEGF actions that primarily drive angiogenic sprouting in response to . In endothelial cells, depletion of intracellular VEGF leads to that cannot be rescued by exogenous VEGF, underscoring the necessity of this autocrine/intracrine loop for baseline endothelial survival. Mechanistically, intracrine VEGF upregulates anti-oxidant genes, including superoxide dismutase 2 (SOD2), via modulation of the FOXO1 , thereby mitigating in cardiac s. It also promotes and function by enhancing , lactate production, and oxygen consumption, preventing mitochondrial fragmentation under stress conditions. In cardiomyocytes, VEGF-B signaling through neuropilin-1 (NRP1) further supports mitochondrial , contributing to maintenance and resilience. These intracrine actions confer protective effects against cardiac stressors, reducing ischemia-reperfusion injury by preserving viable myocardial tissue and limiting apoptosis in both endothelial and cardiomyocyte populations. Intracrine VEGF also enhances coronary collateral formation, bolstering vascular adaptation to chronic ischemia. Evidence from conditional knockouts supports this role; endothelial-specific VEGF deletion (VEGF-ECKO) results in increased cardiac vascular lesions under hypoxia, while cardiomyocyte-specific VEGF reduction diminishes microvascular density and impairs cardiac reserve, heightening vulnerability to injury. Studies from 2014 to 2024, including those on non-canonical VEGF pathways, highlight these protective mechanisms in models of myocardial infarction and hypertrophy.

Therapeutic Implications

Targeting intracrine pathways in cardiovascular diseases offers novel therapeutic strategies by addressing intracellular signaling that conventional extracellular inhibitors may overlook. For the renin-angiotensin system (), intracellular angiotensin II (Ang II) production via noncanonical pathways, such as the Ang-(1-12)/chymase axis, contributes to cardiac remodeling and progression. Chymase inhibitors, which disrupt this intracrine Ang II generation, have shown promise in preclinical models by improving left ventricular function and survival post-myocardial infarction when combined with ACE inhibitors. Similarly, (PTHrP) analogs like PTHrP(1-36) and provide cardioprotection by attenuating in cardiomyocytes subjected to simulated ischemia-reperfusion, potentially mitigating through intracrine modulation of survival pathways. For ischemia, intracrine (VEGF) modulators, including VEGF-B targeted therapies, promote functional recovery in myocardial ischemia by enhancing vascular repair independent of extracellular secretion. Clinical evidence supports the exploration of these approaches, though human trials remain limited as of November 2025. A demonstrated that dehydroepiandrosterone (DHEA) supplementation improved endothelial reactivity (via flow-mediated dilation) and reduced in hypercholesterolemic patients, suggesting benefits for local steroid-mediated cardioprotection, potentially through non-genomic pathways. Standard angiotensin receptor blockers (ARBs), which primarily act extracellularly, have reduced left ventricular mass and in angiotensin II-induced cardiac models, though they may not fully address intracrine signaling. These findings highlight the potential of intracrine-targeted RAS modulation to address residual risks in , where traditional therapies achieve only modest reductions (e.g., 5-18% in cardiovascular mortality). Key challenges in developing intracrine therapeutics include achieving selective delivery across cell membranes without disrupting , as most drugs target extracellular compartments and risk off-target effects on neighboring cells. approaches, such as targeted nanoparticles, are being explored to overcome membrane barriers, but issues like poor and potential in cardiac tissues persist. Additionally, distinguishing intracrine from paracrine effects requires precise biomarkers, complicating clinical translation. Looking ahead, gene therapies targeting cardiac dysfunction show promise for and function restoration in preclinical models as of 2025, including approaches addressing dysregulation in rats that alleviate fibrotic changes. Ongoing preclinical-to-clinical transitions for intracrine modulators, such as components, hold potential to treat resistant by disrupting persistent intracellular signaling and to halt post-myocardial remodeling, reducing adverse ventricular dilation and improving outcomes beyond current standards.

Intracrine in Cancer Biology

Tumor Growth and

Intracrine signaling contributes to tumor growth by enabling cancer cells to autonomously regulate through intracellular ligands that activate receptors without requiring extracellular or stromal interactions. This mechanism allows tumors to independently of external growth factors, enhancing survival in nutrient-poor environments. For instance, intracrine (VEGF) in cells promotes by activating intracellular signaling pathways, including ERK1/2 and AKT phosphorylation, which upregulate cyclins and inhibit ; knockdown of intracellular VEGF reduces cell growth without rescue by exogenous VEGF, confirming its intracrine dependency. In breast cancer, intracrine fibroblast growth factor (FGF) signaling drives proliferation via FGFR activation, leading to downregulation of E-cadherin and nuclear translocation of β-catenin, contributing to cell cycle progression. These pathways highlight how intracrine FGF circumvents the need for paracrine cues from the tumor microenvironment, fostering uncontrolled division. A prominent example is intracrine parathyroid hormone-related protein (PTHrP) in osteosarcoma, where it promotes proliferation via activation of the cAMP-PKA-CREB1 pathway, particularly in p53-deficient cells; this signaling is essential for tumor initiation and maintenance, with PTHrP knockdown reducing cell growth and invasion in vitro and in vivo. Evidence from tumor organoids and biopsies further supports this, as intracrine PTHrP dependency in osteosarcoma organoids correlates with higher Ki-67 proliferation indices, indicating direct links to clinical aggressiveness. This intracrine autonomy confers advantages in cancer, such as resistance to therapies targeting ligand secretion, as internal signaling persists despite inhibitors like ; for example, intracrine VEGF in cells sustains even under treatment. Overall, these mechanisms underscore intracrine signaling's role in fueling tumor , with implications for stroma-independent .

Angiogenesis Promotion

In the context of tumor angiogenesis, intracrine vascular endothelial growth factor (VEGF) plays a critical role by enabling internal signaling, distinct from traditional paracrine mechanisms. Unlike paracrine VEGF gradients that guide broad vascular patterning, intracrine VEGF localizes preferentially to endothelial tip cells at sprout leading edges, facilitating precise invasion into the tumor stroma. Intracrine signaling pathways further amplify by upregulating matrix metalloproteinases (MMPs), such as MMP-2 and MMP-9, which degrade components to enable endothelial invasion and tube formation. Additionally, intracrine VEGF contributes to hypoxia-independent stabilization of hypoxia-inducible factor 1-alpha (HIF-1α) through activation of pathways, sustaining pro-angiogenic even in normoxic tumor regions. Nuclear actions of 2 (FGF2), another key intracrine player, complement these effects by translocating high-molecular-weight isoforms to the , where they potentiate endothelial and resistance to growth-inhibitory conditions, thereby reinforcing vessel network expansion in tumors. These mechanisms profoundly impact tumor progression by sustaining hypoxic cores through persistent , preventing and enabling nutrient delivery to rapidly dividing cancer cells. Intracrine VEGF also enhances by promoting perivascular , where tumor cells exploit newly formed vessels for intravasation and dissemination.

Steroid Hormone Formation

In cancer cells, particularly those of and origin, intracrine steroidogenesis enables the intracellular of circulating precursors, such as (DHEAS), into bioactive hormones like (E2) and (DHT). This process relies on key enzymes, including 17β-hydroxysteroid dehydrogenases (17β-HSDs) for the reduction of to testosterone and subsequent formation of DHT, and for the of androgens to estrogens, all occurring without or reliance on extracellular signaling. This local hormone production is critical for fueling the proliferation of (ER)- and (PR)-positive breast tumors, as well as (AR)-positive prostate cancers, by providing sustained ligand availability for receptor activation. Notably, it allows these tumors to maintain high intratumoral E2 concentrations—often 10- to 50-fold higher than in matched serum—despite systemic reductions in circulating estrogens, thereby evading the suppressive effects of systemic aromatase inhibitors that primarily target peripheral synthesis. Evidence from recent investigations underscores this mechanism; for instance, a 2025 study using liquid chromatography-mass spectrometry analyzed the cell line (among others) and confirmed efficient intracrine conversion of DHEAS to E2 via intermediates like and testosterone. In models, similar pathways sustain DHT levels sufficient for signaling in castration-resistant states, independent of gonadal androgens. The resulting steroids bind and activate nuclear and , promoting their dimerization, DNA binding, and transcription of genes involved in progression and survival, thereby driving tumor growth. Activated receptors also exert feedback on steroidogenic enzymes, such as upregulating expression in breast tissue via ER-mediated mechanisms, which perpetuates the intracrine loop. As a therapeutic target, tissue-selective aromatase inactivators like and its analogs irreversibly bind the within tumor cells, disrupting local E2/DHT synthesis and enhancing against hormone-dependent cancers resistant to non-steroidal inhibitors.

Intracrine in Development and Regeneration

Stem Cell Differentiation

Intracrine signaling plays a pivotal role in directing fate by enabling intracellular retention and autocrine-like activation of ligands and receptors, thereby guiding the exit from pluripotency and commitment to specific lineages without relying solely on extracellular cues. These mechanisms involve loops where factors such as bone morphogenetic proteins (BMPs) and Wnts are internalized or retained within the cell, amplifying lineage-specific during the commitment phase of . For instance, in mesenchymal stem cells (MSCs) is suggested to promote lineage commitment, potentially functioning in an intracrine manner that allows accumulation locally, possibly by unconventional secretion mechanisms, with reductions in BMP2 leading to decreased expression of osteogenic genes like Osterix. In embryonic stem cells (ESCs), local intracrine or autocrine FGF2 signaling maintains pluripotency markers such as OCT4 and NANOG while facilitating transitions to mesodermal lineages upon modulation; knockdown of endogenous FGF2 leads to spontaneous , with exogenous FGF2 supplementation promoting mesoderm-derived cardiovascular progenitors by activating MAPK/ERK pathways. This contrasts with its inhibitory effect on ectodermal fates, as FGF signaling suppresses neural in ESCs, thereby biasing toward mesendodermal commitments. Similarly, intracrine Wnts contribute to pluripotency exit by stabilizing β-catenin intracellularly, though specific intracrine loops are less characterized compared to factors. A prominent example is intracrine VEGF in adult hippocampal neural stem cells (NSCs), where it sustains quiescence and stemness through a cell-autonomous VEGF-VEGFR2 loop localized to the and Golgi apparatus. In these RGL-NSCs, 92.5% co-express Vegfa and Kdr, and disruption via CRISPRi-mediated suppression of VEGFR2 significantly increases and shifts fate toward intermediate progenitors, reducing NSC . This intracrine mechanism is critical during the phase, with studies showing that such loops influence fate decisions in colony models by establishing intracellular gradients that amplify stemness genes like Sox2. Evidence from hippocampal NSC cultures and intact models confirms that blocking intracrine VEGF exhausts the NSC pool, underscoring its role in preventing premature differentiation.

Organogenesis Processes

Intracrine signaling contributes to organogenesis by facilitating precise, cell-autonomous regulation of , , and patterning in embryonic tissues. Similarly, intracrine steroidogenesis generates local gradients of sex steroids, such as testosterone and , that drive development by promoting sex-specific cell fate decisions in primordial germ cells and somatic gonadal cells during early gonadal ridge formation. Mechanisms of intracrine signaling emphasize compartmentalization to avoid diffusion-mediated errors in patterning, allowing signals to interact directly with intracellular receptors or targets. For instance, (PTHrP) exerts intracrine effects in bone by nuclear translocation, sustaining proliferation. Purinergic intracrines, involving intracellular ATP binding to P2X receptors, similarly compartmentalize responses in hematopoietic progenitors, promoting survival. Evidence from model organisms underscores these roles, with mutants disrupting intracrine pathways revealing organ-specific defects; for example, truncation alleles in C (vegfc) impair but may enable intracrine VEGF actions, leading to incomplete intersomitic vessel formation and broader vascular failures during trunk development. A review synthesizes data on purinergic intracrines in hematopoiesis, showing their necessity for primitive erythroid progenitor maturation in the , with disruptions causing anemia-like defects in blood island . Intracrine signaling activity temporally peaks during , when mesendodermal progenitors establish germ layers, and somitogenesis, coordinating oscillatory clock genes for segmental boundaries, ensuring timely axial elongation and positioning. Evolutionarily, intracrine mechanisms are conserved across metazoans, appearing in for foundational patterning; fibroblast growth factors (FGFs) exhibit intracrine translocation in nematodes and arthropods to regulate early embryonic cell fate, a trait retained in vertebrates for analogous roles in tissue specification. This conservation highlights intracrines' ancient utility in preventing signaling noise in compact embryonic environments, from segment polarity to mammalian boundary formation.

Regenerative Medicine Applications

Intracrine signaling has emerged as a promising avenue in by enabling localized control of cellular processes within target tissues, particularly through engineered modifications to enhance tissue repair and therapies. One key approach involves engineering mesenchymal (MSCs) to overexpress intracrines such as (VEGF), which acts intracellularly to promote survival and integration in damaged myocardium. For instance, VEGF-overexpressing MSCs have demonstrated enhanced repair in ischemic heart models by sustaining intracrine loops that boost cell viability and without relying on secretion. Similarly, dehydroepiandrosterone (DHEA) leverages intracrine formation of androgens and estrogens in osteoblasts to stimulate bone regeneration, increasing mineral density and fracture healing in preclinical studies. Evidence from 2020s research supports the application of intracrine (FGF) in cardiac patches, where activates intracellular FGF signaling to mitigate and promote cardiomyocyte in infarcted tissues. These findings build on developmental roles of intracrines in maintenance, adapting them for therapeutic contexts like neural and cardiac regeneration. Despite these advances, challenges persist in intracrine therapies, including the need to tightly control signaling loops to prevent from unchecked intracellular activity, which could lead to uncontrolled proliferation. Delivery remains a hurdle, often addressed via nanoparticles to encapsulate and release intracrines at sites, ensuring targeted uptake while minimizing off-target effects and . Clinical outcomes highlight improved engraftment of engineered stem cells, with intracrine modifications achieving up to 50% retention rates in animal models of cardiac and bone repair, far surpassing unmodified controls. This enhancement supports potential for , where patient-specific intracrines could tailor therapies to individual genetic profiles for optimized regeneration. Recent advances in technologies, as reviewed in 2024, continue to explore applications in , including transcriptional activation for cell fate control that may enhance intracrine pathways in therapies.

Intracrine in Other Physiological Systems

Nervous System Functions

Intracrine signaling plays a critical role in the by enabling localized, cell-autonomous regulation of neuronal maintenance, , and protective responses without reliance on extracellular diffusion. This mechanism allows molecules such as growth factors and hormones to interact with intracellular receptors, supporting the highly polarized architecture of neurons where axons and dendrites require distinct signaling compartments. A prominent example of intracrine signaling in the is (VEGF) in adult hippocampal . In the , radial glia-like neural s (RGL-NSCs) utilize an intracrine VEGF-VEGFR2 loop to maintain quiescence and prevent exhaustive differentiation, ensuring sustained essential for cognitive functions. This process also promotes RGL-NSC proximity to vascular niches, facilitating nutrient access and long-term viability. Another key instance involves intracrine aldosterone in , where lipopolysaccharide-induced production via activates Toll-like receptor 4-dependent innate immune responses, including upregulation of inflammatory mediators through and glycogen synthase kinase-3β pathways. Mechanistically, intracrine-like actions of (BDNF) support growth in adult-born hippocampal neurons by promoting branching and complexity in a cell-autonomous manner, independent of distant sources. Similarly, the intracrine renin-angiotensin system (RAS) modulates through angiotensin II type 2 receptor activation, influencing neuronal differentiation and in brain circuits. These pathways enable precise, localized control within neuronal compartments. Intracrine signaling contributes to during ischemia, as VEGF exerts direct anti-apoptotic effects on neurons preceding vascular changes, reducing infarct size in experimental models. In mood regulation, local intracrine synthesis in the , including neurosteroids like , fine-tunes affective behaviors by modulating and responses in limbic regions. Evidence from mouse models underscores these roles; conditional knockdown of VEGF in RGL-NSCs via lentiviral shRNA disrupts maintenance and vascular association, leading to impaired hippocampal that correlates with deficits in spatial learning and tasks. Human induced pluripotent (iPSC)-derived models have been utilized to study neuronal signaling and disorders, including those involving intracrine mechanisms. The unique advantage of intracrine signaling in polarized neurons lies in its facilitation of compartmentalized responses, allowing independent of axonal versus dendritic domains without cross-talk from secreted ligands.

Metabolic Regulation

Intracrine signaling plays a critical role in regulating cellular by enabling ligands to act within the producing cell, particularly in and glucose homeostasis. In adipocytes, intracrine activation of the free receptor 4 (FFA4, also known as GPR120) at membranes provides a key example of local control over . Upon initiation of , locally released s bind to intracellular FFA4 pools, triggering Gi/o protein-mediated signaling that suppresses further triglyceride breakdown, thus preventing excessive free release. This mechanism establishes a rapid loop at the site of storage, highlighting how intracrine GPCRs sense and modulate intracellular metabolites to maintain metabolic balance. Similarly, local insulin signaling within adipocytes contributes to fine-tuned of handling, where internalized or autocrine insulin pathways enhance and without relying solely on systemic circulation. Mechanistically, intracrine signals often target mitochondria to influence β-oxidation, the primary pathway for . For instance, mitochondrial intracrines such as angiotensin II and transforming growth factor-β (TGF-β), trafficked via megalin receptors, modulate and management, thereby optimizing energy production from lipids. This targeting supports efficient β-oxidation while mitigating in metabolically active tissues like adipose and liver. Additionally, intracrine feedback influences (GLUT) dynamics; in adipocytes, local GPCR signaling, including FFA4, indirectly regulates translocation and activity, promoting glucose influx for synthesis and inhibiting futile cycles of and re-esterification. These processes ensure coordinated and at the cellular level. The physiological impacts of intracrine regulation extend to adipose tissue remodeling and hepatic gluconeogenesis control. In adipose depots, intracrine androgens derived from local biosynthesis promote depot-specific fat distribution and inhibit excessive expansion, contributing to healthier adipose architecture during energy surplus. This remodeling reduces ectopic lipid accumulation and supports insulin sensitivity. In the liver, sex steroids such as androgens can suppress gluconeogenesis, thereby limiting glucose output during fasting or stress states. Evidence from 2025 studies on GPCR intracrines, including FFA4, demonstrates that disruptions—such as in knockout models—lead to dysregulated lipolysis, elevated circulating free fatty acids, and phenotypes resembling metabolic syndrome, including insulin resistance and hyperglycemia. These findings underscore intracrine pathways' relevance to obesity and diabetes, where impaired local signaling exacerbates systemic metabolic dysfunction.

Immune System Roles

Intracrine signaling modulates innate and adaptive immune responses by enabling ligands to act within the producing cell, often amplifying local defense mechanisms without systemic release. In macrophages, the precursor form of interleukin-1α (pro-IL-1α) functions as an intracrine proinflammatory activator, binding intracellularly to induce NF-κB activation and sustain inflammatory gene expression, which in turn influences autophagy pathways to regulate cytokine processing and secretion. Similarly, in glial cells such as astrocytes, Toll-like receptor 4 (TLR4) activation triggers intracrine production of aldosterone via upregulation of steroidogenic enzymes like StAR and CYP11B2, leading to mineralocorticoid receptor (MR)-dependent NF-κB signaling and enhanced expression of complement component C3 and cytokines including IL-1β and TNF-α. This aldosterone-TLR4 cross-talk, demonstrated in lipopolysaccharide (LPS)-stimulated models, promotes neuroinflammation through paracrine effects on neighboring cells while maintaining intracellular homeostasis in glia. These mechanisms contribute to key roles in immune function, such as amplifying and sustaining in . Intracrine vitamin D production in macrophages and dendritic cells, induced by TLR signaling, upregulates like cathelicidin, which enhance phagocytic killing of pathogens and amplify innate responses to . In , intracrine sex steroids in immune cells, such as local estrogen synthesis in synovial macrophages, sustain proinflammatory production (e.g., IL-1β and IL-6) via pathways, contributing to persistent in conditions like . Purinergic signaling further supports these roles through mitochondrial P2X7 receptors in hematopoietic stem/progenitor cells (HSPCs), where intracellular ATP modulates activation to drive sterile and HSPC during immune challenges. Representative examples illustrate these processes in specific immune contexts. Local parathyroid hormone-related protein (PTHrP) exerts intracrine effects in bone-resorbing cells, including osteoclast precursors, where it regulates nuclear localization and gene expression to influence differentiation and survival, indirectly modulating immune-mediated . In hematopoiesis, purinergic intracrines via the complosome—integrating complement and pathways—control HSPC trafficking and innate immune priming, as highlighted in a 2024 review linking these signals to evolutionary immune-hematopoietic rhythms. Recent evidence from 2023–2025 underscores the therapeutic potential of targeting intracrine pathways. Studies show that intracrine inhibitors, such as the MR antagonist , block aldosterone-mediated activation in TLR4-stimulated , reducing production and without broad . Similarly, modulating purinergic complosome activity in HSPCs via inhibitors attenuates excessive responses in innate immunity models. These findings, including 2024 analyses of steroidogenesis in and 2025 explorations of complosome in HSPCs, highlight intracrine targets for mitigating storms in infections or . Such localized interventions offer implications for therapies that avoid systemic side effects, preserving global immune competence while dampening hyperactive responses. For instance, inhibiting intracrine aldosterone in with has shown efficacy in reducing LPS-induced , suggesting applications in neuroinflammatory disorders. Overall, intracrine modulation provides precise control over immune amplification, balancing defense against pathogens and self-tolerance.

References

  1. [1]
    Thirty Years of Intracrinology - PMC - NIH
    In 1984, this laboratory introduced the term intracrine, meaning the action of a peptide hormone within a cell as opposed to acting at cell surface receptors.
  2. [2]
    An intracrine view of sex steroids, immunity, and metabolic regulation
    In strict definition, intracrine signaling is specific to mediators that signal through cytosolic or nuclear receptors, as they can confer signaling effects ...
  3. [3]
    Exploring the Intracrine Functions of VEGF-A - PMC - NIH
    Jan 19, 2021 · This intracellular mode of signalling has been termed “intracrine”, and refers to the direct action of a signalling molecule within the cell ...
  4. [4]
    The mitochondrial component of intracrine action - PubMed - NIH
    In recent years the actions of intracellular-acting, extracellular signaling proteins/peptides (intracrines) have become increasingly defined.
  5. [5]
    The physiological basis of intracrine stem cell regulation - PMC - NIH
    Intracrine peptides and proteins participate in the regulation of adult and pleuripotential embryonic-like stem cells. Included among these factors are VEGF, ...
  6. [6]
    The mitochondrial component of intracrine action - PMC - NIH
    In recent years the actions of intracellular-acting, extracellular signaling proteins/peptides (intracrines) have become increasingly defined.Table 1 · Table 2 · Angiotensin Ii And...
  7. [7]
    Intracrinology - PubMed
    A promising new area in hormone action, namely intracrinology. These steroidogenic and steroid metabolizing enzymes should become a major target of novel ...Missing: intracrine 1984
  8. [8]
    Intracrine - an overview | ScienceDirect Topics
    Intracrine signaling is a mechanism of growth control involving the direct action of growth factors within the cell. Some growth factors produce factor/receptor ...
  9. [9]
    Toward a theory of intracrine hormone action - ScienceDirect.com
    Abstract. A growing body of evidence indicates that in some cases, peptide hormones can function in the intracellular space. These findings are reviewed.Missing: paper | Show results with:paper
  10. [10]
    Cell Signaling - PROGEN Biotechnik GmbH
    The autocrine, juxtacrine, intracrine, paracrine, and endocrine signaling pathway each of which is defined by its individual path from signal molecule to target ...
  11. [11]
    The precursor form of IL-1α is an intracrine proinflammatory activator ...
    The intracellular accumulation of cytokines like IL-1α lacking a signal peptide likely represent an evolutionary advantage, because readily secreted proteins in ...
  12. [12]
    Exploring the Intracrine Functions of VEGF-A - MDPI
    In this review, we describe examples of intracrine VEGF signalling in regulating cell growth, differentiation and survival, both in normal cell homeostasis and ...
  13. [13]
    The Nature of Intracrine Peptide Hormone Action | Hypertension
    The cellular biology of angiotensin: paracrine, autocrine, and intracrine actions in cardiovascular tissues. J Mol Cell Cardiol. 1989;21(suppl 5):63–69. Google ...
  14. [14]
    Intracrinology - ScienceDirect.com
    A promising new area in hormone action, namely intracrinology. These steroidogenic and steroid metabolizing enzymes should become a major target of novel ...
  15. [15]
    The origins of intracrine hormone action - PubMed
    It is here argued that intracrine action is the modern analogue of a biologically ancient mechanism for regulating message translation and ribosome synthesis.Missing: term 1989
  16. [16]
    DHEA and its transformation into androgens and estrogens in ...
    This new field of endocrinology, called intracrinology, describes the local synthesis of androgens and estrogens made locally in each cell of each peripheral ...Missing: shift | Show results with:shift
  17. [17]
    Inhibition of Breast Cancer and Other Roles of Androgens and Their ...
    In a series of animal models, androgens and DHEA have been found to inhibit breast cancer development and growth and to stimulate bone formation.
  18. [18]
    Intracrine Regulation of Estrogen and Other Sex Steroid Levels in ...
    This allows fine-tuning the exposure of responsive tissues and organs to estrogens and other steroids in order to best respond to the physiological needs of ...
  19. [19]
    Dehydroepiandrosterone and Its Metabolite 5-Androstenediol
    All of the clinical studies revealed the safety and positive effects of DHEA treatment on vaginal atrophy parameters, bone mass, and the endogenous DHEA level.
  20. [20]
    Testosterone and Dihydrotestosterone Tissue Levels in Recurrent ...
    Jul 6, 2005 · Median tissue levels of dihydrotestosterone were 91% lower in recurrent prostate cancer (1.25 pmol/g tissue) than AS-BP (13.70 pmol/g tissue; ...Missing: discrepancies endocrinology
  21. [21]
    Join Us at ENDO 2025 - Endocrine Society
    With over 7,000 attendees, nearly 2,500 abstracts and over 200 other sessions, ENDO is the leading global meeting on endocrinology research and clinical care.
  22. [22]
    FGF1 C-terminal domain and phosphorylation regulate intracrine ...
    Feb 4, 2016 · FGF1 nuclear localization is required for these intracellular activities, suggesting that FGF1 regulates p53-dependent apoptosis and neuronal ...
  23. [23]
    Intracrine VEGF Signaling Mediates the Activity of Prosurvival ...
    Multiple studies indicate that VEGF has a direct effect on the survival and growth of a variety of cancer cells (24–28), mostly through an autocrine signaling ...
  24. [24]
    The JAK/STAT signaling pathway: from bench to clinic - Nature
    Nov 26, 2021 · The JAK/STAT pathway constitutes a rapid membrane-to-nucleus signaling module and induces the expression of various critical mediators of cancer and ...Missing: intracrine | Show results with:intracrine
  25. [25]
    The Nature of Intracrine Peptide Hormone Action | Hypertension
    ### Summary of Positive Feedback Loops in Intracrine Signaling for Peptide Hormones like Ang II
  26. [26]
    Interaction between Wnt/β-catenin and RAS-ERK pathways ... - NIH
    Feb 20, 2018 · Aberrant activation of the Wnt/β-catenin and RAS-extracellular signal-regulated kinase (ERK) pathways play important roles in the tumorigenesis ...Missing: intracrine | Show results with:intracrine
  27. [27]
    Angiotensin II Up-Regulates Angiotensin I-Converting Enzyme (ACE ...
    In vitro, Ang II was able to up-regulate ACE and down-regulate ACE2 in human kidney tubular cells, which were blocked by an angiotensin II (AT)1 receptor ...Missing: intracrine | Show results with:intracrine
  28. [28]
    Intracrine sex steroid synthesis and signaling in human epidermal ...
    Nov 12, 2014 · Peripheral intracrine sex steroid synthesis from adrenal precursors dehydroepiandrosterone (DHEA) and DHEA-sulfate has evolved in humans.<|control11|><|separator|>
  29. [29]
    Autocrine loops with positive feedback enable context-dependent ...
    We describe a mechanism for context-dependent cell signaling mediated by autocrine loops with positive feedback. We demonstrate that the composition of the ...
  30. [30]
    Angiotensin II exerts positive feedback on the intrarenal renin ...
    ACE inhibition blocks this positive feedback loop, suggesting that ANG II activates the intrarenal RAS by an ACE-dependent mechanism.Missing: intracrine | Show results with:intracrine
  31. [31]
    [PDF] Quantitative Studies of EGFR Autocrine Induced Cell Signaling and ...
    May 18, 2007 · We demonstrate that positive feedback downstream of the EGF receptor contributes to a 4-fold increase in the ligand release rate of the TGFa.
  32. [32]
    Intracrine angiotensin II functions originate from noncanonical ...
    New research demonstrates that noncanonical mechanisms regulate the synthesis and processing of intracellular angiotensins and brings to light a need to ...
  33. [33]
    Intracellular Angiotensin‐II Interacts With Nuclear ... - PubMed Central
    Intracellular (“intracrine”) angiotensin‐II (Ang‐II) signaling regulates intercellular communication, excitability, and gene expression in cardiomyocytes; ...
  34. [34]
    Angiotensin II-Induced Signal Transduction Mechanisms for Cardiac ...
    While the activation of RAS for a short period has been shown to produce adaptive cardiac hypertrophy for maintaining cardiovascular function, prolonged ...<|separator|>
  35. [35]
    An Insight on Multicentric Signaling of Angiotensin II in ... - Frontiers
    The multifaceted nature of the renin-angiotensin system (RAS) makes it versatile due to its involvement in pathogenesis of the cardiovascular disease.
  36. [36]
    Clinical Relevance of Local Renin Angiotensin Systems - Frontiers
    Local RAS refers to tissue-based mechanisms of Ang peptide formation that operate separately from the circulating RAS.<|control11|><|separator|>
  37. [37]
    Parathyroid Hormone-Related Protein Is Produced in the ...
    Aug 6, 2025 · PTHrP is produced in a wide variety of different cells, including cardiomyocytes. Its production is augmented by mechanical and neurohumoral ...<|separator|>
  38. [38]
    The PTHrP/PTH.1-R Bioregulation System in Cardiac Hypertrophy
    The nuclear import of PTHrP may also occur via an intracrine pathway of a cell expressing PTHrP or in a target cell, as a result of the PTHrP binding to the PTH ...
  39. [39]
    [PDF] PTH-related protein and Type 1 PTH receptor mRNA expression in ...
    PTHrP is expressed in various cells in the cardiovascular system, such as cardiac ... cardiomyocytes become more sensitive to PTHrP ... Intracrine PTHrP protects ...
  40. [40]
    Parathyroid hormone‐related protein and its receptors: nuclear ...
    Jan 29, 2009 · Although the cardiovascular system appears to develop normally in the PTHrP knockout mouse, homologous deletion of the PTH1R results in a higher ...
  41. [41]
  42. [42]
    Parathyroid hormone‐related protein (PTHrP) inhibits mitochondrial ...
    Aug 5, 2025 · Parathyroid hormone‐related protein (PTHrP) inhibits mitochondrial‐dependent apoptosis ... ischemia-reperfusion-induced cardiomyocyte death.
  43. [43]
    Parathyroid hormone-related peptide protects cardiomyocytes from ...
    PTHrP reportedly improves resistance against apoptosis ... demonstrated that the suppressing the activation of ERK1/2 inhibits apoptosis of cardiomyocytes ...
  44. [44]
    Parathyroid Hormone-Related Peptide and Its Analog ...
    Apr 19, 2022 · Two published reports provide indirect support for the concept that PTHrP may render cardiomyocytes resistant to ischemia-reperfusion-induced ...
  45. [45]
    Parathyroid hormone-related peptide improves contractile ... - PubMed
    Parathyroid hormone-related peptide (PTHrP) was found to improve contractile function of stunned myocardium in pigs. The peptide is released from coronary ...Missing: vascular | Show results with:vascular
  46. [46]
    Cardiac-specific effects of parathyroid hormone-related peptide
    Objective: Parathyroid hormone-related peptide (PTHrP) improves heart function of post-ischemic and stunned myocardium and is released from the heart under ...
  47. [47]
    Parathyroid hormone-related protein and its receptors
    In a more recent study, a second tetrabasic KKKK(147 – 150) motif has been proven to determine intracrine regulatory effects of PTHrP(1 – 173) in human ...
  48. [48]
    Midgestational lethality in mice lacking the parathyroid hormone ...
    PTHrP is a key developmental regulatory protein and a potent vasoactive agent ... Mice, Knockout; Microscopy, Electron; Myocardium / chemistry; Myocardium ...Missing: impaired | Show results with:impaired
  49. [49]
    Midgestational Lethality in Mice Lacking the Parathyroid Hormone ...
    Although the cardiovascular system appears to develop normally in the PTHrP knockout mouse ... The Na+-Ca2+ exchanger is essential for embryonic heart development ...
  50. [50]
    Autocrine VEGF maintains endothelial survival through regulation of ...
    Highlighted Article: Intracellular VEGF signaling in endothelial cells regulates mitochondria function and levels of FOXO1. ... intracrine VEGF signaling ...
  51. [51]
    Cardiotoxicity with vascular endothelial growth factor inhibitor therapy
    May 8, 2018 · In fact, in the absence of auto-/intracrine VEGF, paracrine sources are not sufficient to sustain endothelial cell survival. Of further ...
  52. [52]
    The Role of the VEGF Family in Coronary Heart Disease - Frontiers
    The VEGF family can also prevent AS by promoting angiogenesis and regulating oxidative stress. In terms of angiogenesis, coronary artery AS can lead to ischemia ...
  53. [53]
    A cardiac myocyte vascular endothelial growth factor paracrine ... - NIH
    Thus, cardiac myocyte-specific deletion of VEGF results in a dilated thin-walled adult heart. Although this morphometric phenotype is consistent with that of an ...
  54. [54]
    Novel Cardiac Intracrine Mechanisms Based on Ang-(1-12 ...
    Targeting intracellular RAS signaling may improve current therapies aimed at reducing the burden of heart failure. Keywords: intracrine, angiotensin-(1-12), ...
  55. [55]
    Vascular endothelial growth factor-B: Impact on physiology and ...
    This uniqueness of VEGF-B warrant further cogitation of the therapeutic potential of VEGF-B for promoting functional recovery of myocardial ischemia.Treating Diabetes By... · Vegf-B And Lipids... · Targeted Vegf-B Therapy For...
  56. [56]
    In Vivo and In Vitro Evidences of Dehydroepiandrosterone ...
    Nov 2, 2025 · The identification of the beneficial cardiovascular effects of DHEA (S) and a better understanding of the involved mechanisms should be helpful ...Missing: intracrine 2020s
  57. [57]
    Comparative effects of DHEA vs. testosterone, dihydrotestosterone ...
    Hormonal effects on steroid receptor levels included decreases in AR mRNA of 80% with DHT and T treatments and of 70–75% with DHEA and E2 administration, with ...Cell Proliferation Assays · Hormone Treatments For Gene... · Igf-Ir Mrna<|separator|>
  58. [58]
    The Grand Challenges in Cardiovascular Drug Delivery - Frontiers
    One of the Grand Challenges in medicine in general, and cardiovascular drug delivery in particular, is the need of personalized therapies. Currently, medicine ...
  59. [59]
    Advanced Nanomedicine Delivery Systems for Cardiovascular ...
    Conventional therapies for CVDs often grapple with challenges such as inadequate targeting precision, suboptimal therapeutic efficacy, and potential adverse ...
  60. [60]
  61. [61]
  62. [62]
  63. [63]
    Autocrine epidermal growth factor signaling stimulates directionally ...
    Presentation in the intracrine (sEGF) mode resulted in a signaling loop that stimulated cell proliferation, but in a structurally disorganized fashion. This ...
  64. [64]
  65. [65]
    Intracellular and intercellular signaling networks in cancer initiation ...
    Intracellular and intercellular signaling networks in cancer initiation, development and precision anti-cancer therapy. RAS acts as contextual signaling hub.
  66. [66]
  67. [67]
  68. [68]
  69. [69]
    Intracrine Formation of Steroid Hormones in Breast Cancer ... - MDPI
    It is crucial to note that serum hormone concentrations may not reflect local hormonal activities in target tissues, emphasizing the importance of studying ...
  70. [70]
    Intracrine androgen metabolism in prostate cancer progression - NIH
    Intracrine androgen metabolism in prostate cancer progression: mechanisms of castration resistance and therapeutic implications ... 1989;68(2):461–468. doi ...
  71. [71]
    Estrogens in the breast tissue: a systematic review - PMC
    The levels of estrone (E1) and estradiol (E2) measured in breast tumor tissue from postmenopausal women were markedly higher than their levels in plasma, and ...
  72. [72]
    Steroidogenesis in castration-resistant prostate cancer - ScienceDirect
    Castration resistance is in part attributable to aberrant activation of androgen receptor (AR) signaling by the intracrine activation of androgen precursors ...
  73. [73]
    Steroidogenesis in castration-resistant prostate cancer - PubMed
    Nov 11, 2022 · These steroidogenesis pathways produce potent androgens that promote tumor resistance to endocrine therapy including novel ARPIs.
  74. [74]
    Sex steroid-producing enzymes in human breast cancer in
    Dec 1, 2005 · Breast carcinoma tissues have been demonstrated to process intracrine activity. Locally produced biologically active estrogens act in breast ...
  75. [75]
    Aromatase Inhibition: Translation into a Successful Therapeutic ...
    Whereas first- and second-generation aromatase inhibitors inhibit estrogen synthesis in vivo by up to 90%, the third-generation compounds anastrozole, ...
  76. [76]
    Bmp2 in osteoblasts of periosteum and trabecular bone links ... - NIH
    Primarily, we propose that Bmp2ob stimulates endogenous Bmp2msc, which collectively drives the MSC to osteoblast differentiation pathway in an intracrine or ...
  77. [77]
    A Complex Role for FGF-2 in Self-Renewal, Survival, and Adhesion ...
    Human ESCs express high levels of endogenous FGF-2 that might signal in autocrine, paracrine, or intracrine ways. To address the effects of intrinsic FGF-2 ...
  78. [78]
    FGF signalling inhibits neural induction in human embryonic stem cells
    Nov 15, 2011 · Our study assigns a novel, biphasic role to FGF/ERK signalling in the neural induction of hESCs, which may also have utility for applications requiring the ...
  79. [79]
    Intracrine VEGF Signaling Is Required for Adult Hippocampal Neural ...
    Mar 25, 2025 · We identified a VEGF-VEGFR2 intracrine signaling mechanism within adult DG NSCs that prevents NSC exhaustion and supports their proximity to local blood ...
  80. [80]
    Sonic Hedgehog Gene Delivery to the Rodent Heart Promotes ...
    Jan 5, 2010 · ... Shh. The anticipated objective of our multipronged strategy was to achieve intracrine, autocrine, and paracrine effects of Shh protein which ...
  81. [81]
    Minireview: Parathyroid Hormone-Related Protein as an Intracrine ...
    ... intracrine fashion. This review discusses the mechanisms by which PTHrP may gain access to the nucleus/nucleolus and the functional consequences of this ...
  82. [82]
    PTHrP and skeletal development - PubMed
    Parathyroid hormone-related protein (PTHrP) participates in the regulation of endochondral bone development. After the cartilage mold is established in ...
  83. [83]
    Hematopoiesis Revolves Around the Primordial Evolutional Rhythm ...
    Feb 16, 2024 · This review will focus on the data that purinergic signaling and innate immunity carry on their ancient developmental task in hematopoiesis.
  84. [84]
    A truncation allele in vascular endothelial growth factor c reveals ...
    An intracrine mechanism might also explain differences in ISV defects between vegfcum18 mutants and other examples of vegfc deficiency in zebrafish, which can ...
  85. [85]
    Fibroblast growth factors: from molecular evolution to roles in ... - NIH
    The FGF-signalling system has been conserved throughout metazoan evolution. Two Fgf-like genes, egl-17 and let-756, have been identified in the nematode ...
  86. [86]
    Vascular endothelial growth factor expressing mesenchymal stem ...
    Our results showed that transplanted VEGF expressing MSCs significantly improved chronic myocardial ischaemia as documented by NOGA mapping, improved collateral ...Vegf Gene Transfer And... · Results · Vegf Expression Of Mscsin...
  87. [87]
    VEGF improves survival of mesenchymal stem cells in infarcted hearts
    Aug 6, 2025 · In conclusion, VEGF protects MSCs from culture-induce cellular stress and improves their viability in ischemic myocardium, which results in ...Missing: intracrines | Show results with:intracrines
  88. [88]
    DHEA and the intracrine formation of androgens and estrogens in ...
    We have demonstrated for the first time a series of medically important beneficial effects of DHEA administered for 12 months to post-menopausal women.Missing: prasterone | Show results with:prasterone
  89. [89]
    Recent Advances in Gene Therapy for Cardiac Tissue Regeneration
    FGF signaling can influence the whole process of angiogenesis and cardiomyocyte mitosis. Activation of FGFR1 or FGFR2 has been demonstrated to have a positive ...
  90. [90]
    The Multifunctional Contribution of FGF Signaling to Cardiac ...
    The differentiation of SHF progenitors from human pluripotent stem cells, which can later generate cardiac fibroblasts, is attributed to the effect of FGF2 as ...
  91. [91]
    Intrarenal Generation of Aldosterone Contributes to Ischemia ...
    While Aldo is believed to predominantly originate from the adrenal glands, this study provides evidence to support the involvement of intrarenal ...
  92. [92]
  93. [93]
    Challenges in Development of Nanoparticle-Based Therapeutics - NIH
    This review summarizes challenges likely to be encountered during the development and approval of nanoparticle-based therapeutics.
  94. [94]
    Targeted therapy in chronic diseases using nanomaterial-based ...
    Aug 30, 2019 · This review aims to cover various nanomaterial-based drug delivery systems, their applications in the diagnosis and therapeutics of chronic diseases, and other ...
  95. [95]
    Assessment and Optimization of Cell Engraftment after ... - NIH
    Of the cells that are retained in the heart 24 hrs after delivery, a small percentage (from <1 to 20% depending on the cell type) will stably engraft as ...Missing: intracrine | Show results with:intracrine
  96. [96]
    Engraftment Patterns of Human Adult Mesenchymal Stem Cells ...
    Therefore, this study aimed to investigate the effects of engraftment characteristics of MSCs (ie, different numbers and distribution patterns) on arrhythmicity ...Missing: intracrine | Show results with:intracrine
  97. [97]
    The Interface of Gene Editing with Regenerative Medicine - PMC
    Nov 30, 2024 · In this review, we explore the progress and future prospects of CRISPR technologies in regenerative medicine, focusing on how gene editing has ...Missing: intracrine | Show results with:intracrine
  98. [98]
    CRISPR technologies: Revolutionizing regenerative medicine
    Feb 27, 2025 · News Release 27-Feb-2025. CRISPR technologies: Revolutionizing regenerative medicine. Peer-Reviewed Publication. Higher Education Press.Missing: intracrine synthesis
  99. [99]
    (PDF) High-fidelity reprogramming into Leydig-like cells by CRISPR ...
    Aug 9, 2025 · Using a combination of CRISPR activation and paracrine factors, the fibroblasts have been reprogramed into Leydig-like cells (LLCs). The ...
  100. [100]
    Internalized TSH receptors en route to the TGN induce local G s
    Sep 5, 2017 · In this study, we address these important questions by following the internalization/trafficking of an endogenous receptor in real time, using ...
  101. [101]
    Toll‐like receptor 4‐dependent innate immune responses are ...
    Jun 28, 2024 · We conclude that intracrine aldosterone may be involved in the TLR4-dependent innate immune responses of astrocytes and can trigger paracrine effects.
  102. [102]
    Autocrine Action of BDNF on Dendrite Development of Adult-Born ...
    Jun 3, 2015 · Furthermore, we found that elevated dendrite growth in these neurons caused by voluntary exercise also depends on autocrine BDNF action.Missing: nuclear intracrine
  103. [103]
    Physiopathology of the Brain Renin-Angiotensin System - MDPI
    AT2R plays a crucial role in neuronal differentiation and regeneration, as well as in synaptic plasticity and brain development [14,15]. Its activation is ...
  104. [104]
    Beyond vessels: unraveling the impact of VEGFs on neuronal ...
    Mar 6, 2025 · VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells.
  105. [105]
    Steroid Transport, Local Synthesis, and Signaling within the Brain
    This sustained synthesis of steroids may regulate several brain functions and participate to brain homeostasis, neurotransmission, neurogenesis, and brain ...Missing: intracrine mood
  106. [106]
    Vascular endothelial growth factor regulates adult hippocampal cell ...
    VEGF is also required for the increased cell proliferation and neurogenesis that occurs after adult mice are exposed to environmental enrichment (Cao et al., ...Missing: intracrine | Show results with:intracrine
  107. [107]
    Multiomics Evaluation of Human iPSCs and iPSC-Derived Neurons
    Feb 28, 2024 · Human induced pluripotent stem cells (iPSCs) can be differentiated into neurons, providing living human neurons to model brain diseases.
  108. [108]
    Mechanisms of Polarized Organelle Distribution in Neurons - Frontiers
    Mar 30, 2016 · Neurons are highly polarized cells exhibiting axonal and somatodendritic domains with distinct complements of cytoplasmic organelles.Missing: intracrine | Show results with:intracrine
  109. [109]
    Intracrine FFA4 signaling controls lipolysis at lipid droplets - Nature
    Aug 5, 2025 · A prime example of a tightly regulated metabolic pathway is lipolysis, stimulated by hormones like adrenaline acting upon Gs-coupled receptors ...
  110. [110]
    Insulin action in adipocytes, adipose remodeling, and systemic effects
    A growing body of evidence documents how adipocytes, in response to insulin, contribute to the control of whole-body nutrient homeostasis.Missing: intracrine | Show results with:intracrine
  111. [111]
    Megalin-Mediated Trafficking of Mitochondrial Intracrines
    Megalin shuttles mitochondrial intracrines (angiotensin II, stanniocalcin-1 and TGF-ß) from the cell surface to the mitochondria through the retrograde early ...
  112. [112]
    Intracrine androgen biosynthesis, metabolism and action revisited
    Hammond G.L. Plasma steroid-binding proteins: primary gatekeepers of steroid hormone action. ... The intracrine sex steroid biosynthesis pathways. Prog. Brain Res ...
  113. [113]
    An intracrine view of sex steroids, immunity, and metabolic regulation
    Sex steroids signal through nuclear receptors, and a membrane receptor (GPR30) also has been identified for estrogen signaling. Therefore, intracrine function ...
  114. [114]
    Regulation of intracrine production of 1,25-dihydroxyvitamin D and ...
    Jul 1, 2012 · Regulation of intracrine production of 1,25-dihydroxyvitamin D and its role in innate immune defense against infection · Author · Affiliation.
  115. [115]
    C-Terminal Region of PTHrP, in Addition to the Nuclear Localization ...
    It has been suggested that the proliferative effects of PTHrP, mediated by the nuclear entry and action of PTHrP in an intracrine manner, may participate in the ...Materials And Methods · Pthrp Immunoradiometric... · Results
  116. [116]
    Expression of innate immunity genes in human hematopoietic stem ...
    Mar 19, 2025 · Thus, our data sheds new light on complosome as a novel regulator of hematopoiesis that involves intracrine activation of the C5a-C5aR-Nlrp3 ...
  117. [117]
    Hematopoietic stem cells on the crossroad between purinergic ...
    May 15, 2023 · In this review, we will discuss the coordinated effects of these pathways in regulating the biology of HSPC. Importantly, both purinergic ...