Fact-checked by Grok 2 weeks ago

Hepatic stellate cell

Hepatic stellate cells (HSCs), also known as cells or fat-storing cells, are liver-specific mesenchymal located in the perisinusoidal space of Disse between sinusoidal endothelial cells and hepatocytes, comprising about 5–15% of total liver cells and playing essential roles in storage, (ECM) homeostasis, and in their quiescent state. In response to hepatic injury from toxins, viruses, , or metabolic , quiescent HSCs undergo activation, transitioning into proliferative, contractile myofibroblast-like cells that lose vitamin A droplets, express α-smooth muscle actin (α-SMA), and secrete excessive ECM components like collagen type I, thereby driving as a central mechanism in chronic liver diseases ranging from to and (HCC). First described as "star cells" by von Kupffer in and formally named by in 1951 for their lipid-laden appearance, HSCs were isolated and characterized in the , with their nomenclature standardized as hepatic stellate cells in 1996 due to their stellate morphology and multifunctional nature. In the quiescent state, HSCs exhibit a spindle-shaped with dendritic processes, an oval , and prominent cytoplasmic droplets containing 80–90% of the body's retinyl esters, supported by ultrastructural features like rough and a Golgi complex for metabolism and ECM synthesis. They maintain liver architecture by regulating ECM turnover, supporting sinusoidal endothelial fenestrations via paracrine signals, and contributing to through interactions with regulatory T cells and myeloid-derived suppressor cells, while also secreting s such as hepatocyte (HGF) and (VEGF) to promote proliferation during regeneration. Metabolically, quiescent HSCs rely on pathways for storage and low-level , ensuring energy and nutrient without . Activation of HSCs is a dynamic, two-phase process initiated by liver damage signals like (ROS), proinflammatory cytokines (e.g., TGF-β and PDGF), and ligands, leading to loss of lipid droplets, , and reprogramming toward , glutaminolysis, and oxidation to fuel proliferation and migration. Recent has revealed HSC heterogeneity, with zonal subpopulations (e.g., portal vs. central vein) exhibiting distinct pro-regenerative or profibrogenic potentials, and reversible inactivation upon injury resolution through mechanisms like and signaling. In , activated HSCs act as key effectors of fibrogenesis by cross-talking with macrophages and hepatocytes, exacerbating inflammation in conditions like nonalcoholic steatohepatitis (NASH) and promoting HCC progression via remodeling and , though certain HSC subsets may exert protective effects through HGF-mediated tumor suppression. Their central role positions HSCs as promising therapeutic targets, with ongoing research exploring antifibrotic strategies like FXR agonists and inhibitors to halt activation and reverse .

Anatomy

Location and distribution

Hepatic stellate cells (HSCs) are primarily located in the perisinusoidal of Disse, a narrow extracellular compartment situated between the basolateral surfaces of hepatocytes and the abluminal side of sinusoidal endothelial cells throughout the liver lobule. This positioning allows HSCs to form intimate associations with the hepatic microvasculature and parenchymal cells, facilitating their roles in maintaining liver architecture. In terms of distribution, HSCs constitute approximately 5-10% of the total resident cell population in the liver, with a heterogeneous pattern across the lobular zones. They exhibit higher density in the pericentral (zone 3) and midzonal regions compared to the periportal (zone 1) areas, particularly in models where A-storing lipid droplets—a hallmark of quiescent HSCs—are more abundant in perivenular and midzonal locations. In humans, a slight predominance is observed in the pericentral zone, with single-cell analyses revealing distinct subpopulations such as central vein-associated HSCs enriched near zone 3 and portal vein-associated HSCs closer to zone 1, reflecting zonal heterogeneity in and positioning. HSCs interact closely with neighboring cells through elongated cytoplasmic extensions that extend along sinusoids, enabling direct apposition to sinusoidal endothelial cells, hepatocytes, and Kupffer cells. These processes support bidirectional communication and within the sinusoidal niche, without extending into the sinusoidal .

Cellular morphology

Hepatic stellate cells (HSCs) in their quiescent state display a distinctive star-shaped , featuring a spindle-shaped body with long, dendritic cytoplasmic processes that extend along the liver sinusoids in the perisinusoidal space of Disse. These processes often include thorny microprojections that facilitate interactions with neighboring cells. A hallmark of quiescent HSCs is the presence of abundant droplets, which occupy 20-30% of the cell volume and primarily consist of retinyl esters, storing 80-90% of the total retinoids in the liver. These lipid droplets can be visualized through techniques such as electron microscopy, which reveals their subendothelial location, or gold chloride staining, which highlights their content. Key immunohistochemical markers for identifying quiescent HSCs include desmin, an protein expressed in both and cells; (GFAP), which is strongly associated with the quiescent phenotype and early activation stages; and cytoglobin, a family member particularly prominent in HSCs. Additionally, , an , is expressed in quiescent HSCs and serves as a specific marker to distinguish them from other liver mesenchymal cells and myofibroblasts, as it is absent in portal fibroblasts and periductal myofibroblasts. Upon , HSCs undergo a profound morphological transformation, losing their lipid droplets and adopting a flattened, myofibroblast-like appearance with an enlarged body and reduced or altered cytoplasmic processes. This state is characterized by the proliferation of rough (rER) and Golgi apparatus, supporting increased protein synthesis for components, along with the formation of contractile bundles. Activated HSCs robustly express α-smooth muscle actin (α-SMA), a reliable marker of their into contractile, fibrogenic cells. Ultrastructurally, quiescent HSCs possess an oval or elongated , often compressed by adjacent vacuoles and containing indicative of transcriptional activity, alongside mitochondria for energy production and peroxisomes involved in . In the activated form, the may appear more ruffled, with enhanced development reflecting metabolic shifts, though storage structures are notably diminished.

Physiology

Quiescent state functions

In the quiescent state, hepatic stellate cells (HSCs) serve as the primary reservoir for in the body, storing 50-80% of total retinoids primarily as esters within cytoplasmic lipid droplets. This storage is tightly regulated by retinoid-associated proteins, including cellular retinol-binding protein type I (CRBP-I) and lecithin:retinol acyltransferase (LRAT), which facilitate retinol uptake, esterification, and mobilization to maintain systemic . These cells esterify incoming retinol from dietary sources or circulation, preventing while ensuring availability for extrahepatic needs, such as and immune . Quiescent HSCs also contribute to extracellular (ECM) homeostasis by producing low levels of basement membrane components, including type IV and , which support sinusoidal structure and endothelial integrity. This synthesis is balanced by expression of matrix metalloproteinases (MMPs), such as MMP-2 and MMP-9, and their inhibitors (TIMPs), enabling controlled turnover to prevent excessive deposition or degradation in the healthy liver. Such regulated ECM dynamics maintain vascular patency and facilitate nutrient exchange between hepatocytes and blood. Quiescent HSCs support by secreting growth factors such as hepatocyte growth factor (HGF) and (VEGF), which promote and , respectively. Furthermore, quiescent HSCs regulate liver zonation through of R-spondin 3 (RSPO3), a key activator of Wnt/β-catenin signaling that establishes metabolic gradients across hepatic lobules. RSPO3 from HSCs amplifies Wnt ligand activity, promoting periportal for and zonated expression of metabolic enzymes like in pericentral regions. This paracrine mechanism ensures patterned functions essential for overall and . In terms of immune modulation, quiescent HSCs function as liver-resident antigen-presenting cells (APCs), particularly for natural killer T (NKT) cells, by expressing and CD1d to present glycolipids and support NKT proliferation via IL-15 production. They also secrete anti-inflammatory cytokines such as IL-10, fostering by suppressing pro-inflammatory T cell responses and promoting activity in the hepatic microenvironment. This dual role helps maintain in the liver, preventing unwarranted during steady-state conditions.

Activation mechanisms

Hepatic stellate cells (HSCs) undergo in response to , transitioning from a quiescent state characterized by storage to a proliferative, fibrogenic myofibroblast-like . This process is primarily triggered by paracrine signals from damaged s and activated Kupffer cells, including transforming growth factor-β1 (TGF-β1), (PDGF), and (ROS). TGF-β1, secreted by injured hepatocytes, initiates early events, while PDGF from Kupffer cells and endothelial cells promotes and survival of HSCs. ROS, generated during from hepatocyte damage, further amplifies these signals by activating redox-sensitive pathways in HSCs. The transdifferentiation process involves several hallmark changes: HSCs lose their retinoid (vitamin A) droplets, a feature of their quiescent state, and begin to proliferate through PDGF receptor signaling, which stimulates cell cycle progression via mitogen-activated protein kinase (MAPK) pathways. Concurrently, there is upregulation of α-smooth muscle actin (α-SMA) and other contractility proteins, such as caldesmon and calponin, marking the shift to a myofibroblast phenotype capable of exerting contractile forces on the extracellular matrix. This proliferation is dose-dependent on PDGF isoforms, particularly PDGF-BB, and is essential for expanding the HSC population during chronic injury. Key signaling pathways orchestrate this activation. The canonical TGF-β/SMAD pathway is central to fibrogenic , where TGF-β1 binds to its receptors, leading to of SMAD2 and SMAD3, which translocate to the nucleus to induce transcription of type I and other extracellular matrix components. Epigenetic modifications, including histone acetylation and , promote the myofibroblast phenotype by altering chromatin accessibility; for instance, increased activity sustains profibrotic during prolonged activation. These changes are reinforced by interactions with altered stiffness, creating a feed-forward loop. Despite the progressive nature of , HSCs retain some plasticity, with potential for partial reversion to a quiescent-like state under resolving conditions. PPARγ agonists, such as , can induce phenotypic reversal by enhancing and downregulating fibrogenic genes, as demonstrated in experimental models of liver . Additionally, interactions with natural killer () cells provide resolution signals, where NK cell-derived interferon-γ promotes HSC , limiting excessive during repair. This reversion capacity underscores the therapeutic potential of targeting these mechanisms to halt progression.

Pathophysiology

Role in liver fibrosis

Activated hepatic stellate cells (HSCs) play a central role in driving liver by undergoing into myofibroblast-like cells, which excessively deposit (ECM) components and disrupt normal matrix . This process is triggered by profibrogenic signals such as transforming growth factor-β (TGF-β), leading to the accumulation of in response to chronic liver injury. The imbalance between ECM synthesis and degradation by these cells results in progressive , potentially advancing to if unresolved. A hallmark of activated HSCs is their heightened production of ECM proteins, including fibrillar collagens types I and III, , and proteoglycans, which constitute up to 80% of the total fibrillar in fibrotic livers. This overproduction creates a dense, stiff matrix in the space of Disse, favoring net deposition over physiological turnover and perpetuating fibrogenic signaling through mechanotransduction pathways. Concurrently, activated HSCs upregulate tissue inhibitors of metalloproteinases (TIMPs), particularly TIMP-1, which potently suppress the activity of matrix metalloproteinases (MMPs) such as MMP-2 and MMP-9, thereby inhibiting ECM degradation and exacerbating matrix accumulation. In addition to ECM modulation, activated HSCs acquire contractile properties through the expression of α-smooth muscle actin (α-SMA), enabling them to constrict and remodel the sinusoidal architecture. This α-SMA-mediated contraction promotes sinusoidal capillarization, where fenestrae in liver sinusoidal endothelial cells are lost, impairing blood flow and contributing to elevated portal pressure characteristic of advanced . As a regulatory mechanism, activated HSCs can enter , which acts as a brake on excessive by limiting their proliferative and fibrogenic potential. Senescent HSCs upregulate surface ligands such as and MICB, which bind to the receptor on natural killer (NK) cells, recruiting these immune cells to selectively eliminate the senescent HSCs and thereby attenuate progression. Recent studies as of 2024 have explored inducing HSC senescence via targeted delivery of cGAS-STING activators to enhance immune clearance and reverse .

Involvement in other liver diseases

Hepatic stellate cells (HSCs) play a multifaceted role in hepatocellular carcinoma (HCC), primarily as the main source of cancer-associated fibroblasts within the tumor microenvironment, where they foster a desmoplastic stroma that supports tumor progression. Activated HSCs promote angiogenesis by secreting vascular endothelial growth factor A (VEGFA), facilitating tumor vascularization and nutrient supply essential for HCC growth. They also contribute to immune evasion through expression of programmed death-ligand 1 (PD-L1), which suppresses antitumor immune responses by inhibiting T-cell activity. Furthermore, HSCs aid metastasis by remodeling the extracellular matrix and secreting factors like stromal cell-derived factor 1 (SDF-1) via CXCR4 signaling, creating a permissive niche for tumor cell invasion and dissemination. In liver regeneration, activated HSCs transiently aid repair by releasing HGF and cytokines like (PDGF) and transforming growth factor-β (TGF-β), promoting mitosis and vascular remodeling within the first week of recovery. However, prolonged activation risks fibrotic scarring through excessive deposition, potentially distorting liver architecture if resolution via or fails; recent findings as of 2024 highlight PFKFB3-mediated in HSCs as a key driver of regenerative proliferation with fibrogenic potential. In alcoholic and metabolic dysfunction-associated (MASH), HSCs exacerbate disease progression by responding to lipotoxic signals, including products like oxidized low-density lipoproteins, which upregulate (TLR4) and sensitize cells to TGF-β via suppression of and activin membrane-bound inhibitor (). Activated HSCs release proinflammatory such as , , IL-8, and , recruiting macrophages and amplifying through p38 MAPK signaling and stellakine production. This milieu, combined with and PDGF pathways, drives sustained differentiation and I/III deposition, accelerating transition from to advanced and . in HSCs further sustains activation by facilitating lipid release and profibrogenic signaling, underscoring their central role in metabolic outcomes. During viral hepatitis caused by hepatitis B virus (HBV) or (HCV), HSCs contribute to chronic inflammation by directly responding to viral signals, such as HBV surface antigen (HBsAg) and X protein (HBx), which activate TLR/MyD88/ pathways leading to increased expression of α-smooth muscle actin (α-SMA), collagen I (COL1A1), and PDGF-B. Paracrine factors from infected hepatocytes, including TGF-β (induced by HBx/Egr-1) and (HMGB1) via , further perpetuate HSC activation and ECM production. In HBV, HBx inhibits ferroptosis by binding PPARγ and promotes Th17 cell recruitment via IL-17/, fostering a profibrotic inflammatory that transitions to . For HCV, core protein and exosomes containing miR-222 enhance HSC migration and contraction, while co-infection with HBV amplifies TGF-β1-mediated OCT4/Nanog signaling, driving chronic inflammation and fibrogenesis.

History and Research

Discovery and nomenclature

The discovery of hepatic stellate cells (HSCs) dates back to the late , when anatomist Karl Wilhelm von Kupffer first observed star-shaped perisinusoidal cells in the liver using gold chloride staining techniques originally intended to visualize neural elements. These cells, termed "Sternzellen" (star cells), were initially described as residing in the space of Disse and were thought to be part of the liver's framework, though von Kupffer later contributed to the confusion by associating similar structures with the phagocytic cells now known as Kupffer cells (liver macrophages). Over the subsequent decades, these observations were largely overlooked or misinterpreted, with the cells occasionally referenced in histological studies but not distinctly characterized. A pivotal advancement occurred in 1951, when Japanese anatomist Toshio Ito refined the gold chloride staining method to clearly delineate -laden stellate cells in the livers of vitamin A-supplemented rats, highlighting their perisinusoidal location and cytoplasmic extensions that ensheath sinusoidal endothelial cells. Ito's work emphasized the cells' prominent droplets, which became more evident under conditions of high intake, distinguishing them from other liver cell types and reviving interest in their identity. This staining approach allowed for the first reliable visualization of these cells in mammalian livers, marking a key milestone in their identification as a unique population. The nomenclature of these cells evolved through the mid-20th century, reflecting their morphological and functional attributes: Ito's descriptions led to the term "Ito cells," while their lipid content prompted names like "fat-storing cells," "lipocytes," or "perisinusoidal cells," and some early works likened them to due to their encircling processes. In 1971, Keith Wake unified these concepts by demonstrating through that von Kupffer's Sternzellen, Ito's fat-storing cells, and other perisinusoidal descriptions referred to the same , further establishing their role in storage via that revealed autofluorescent retinoid droplets comprising up to 80% of the body's total reserves. By the 1980s, as isolation techniques advanced and their broader physiological significance emerged in , the term "hepatic stellate cells" (HSCs) gained prominence to encapsulate their star-like morphology and hepatic specificity. The nomenclature was formally standardized as "hepatic stellate cells" in 1996.

Recent advances

Recent advances in hepatic stellate cell (HSC) research, particularly through single-cell RNA sequencing (scRNA-seq), have unveiled significant heterogeneity among these cells, identifying distinct subpopulations such as lipid-rich quiescent HSCs and matrix-producing activated . These studies reveal spatiotemporal diversity, with peri-central vein HSCs emerging as primary contributors to myofibroblast formation during injury. Furthermore, a 2024 single-cell atlas demonstrates conserved activation trajectories across various liver injuries, including chemical and metabolic insults, highlighting shared transcriptional programs that drive fibrogenesis regardless of . Emerging functions of HSCs extend beyond to include of liver size, zonation, and via the RSPO3-Wnt/β-catenin signaling axis, as elucidated in a 2025 study showing HSC-derived RSPO3 modulates proliferation and metabolic gradients. This pathway maintains liver by enhancing Wnt receptor expression on hepatocytes, with disruptions linked to impaired regeneration and progression. Additionally, HSCs engage in exosome-mediated intercellular communication, transferring miRNAs and proteins to neighboring cells like macrophages and hepatocytes, which influences both homeostatic and disease states such as in non-alcoholic (NASH). For instance, HSC-derived exosomes promote pro-fibrotic release from macrophages, amplifying deposition. Therapeutic strategies targeting HSCs have advanced with HSC-specific inhibitors and agonists showing promise in reversing . FXR agonists, such as (OCA) and the non-steroidal INT-787, inhibit HSC activation by upregulating matrix-degrading enzymes like RECK, reducing and inflammation in models. Senolytics, including and combinations, selectively clear senescent activated HSCs in preclinical and (HCC) models, alleviating and tumor progression by disrupting senescence-associated secretory phenotypes. Gliotoxin, a fungal , targets activated HSCs via selective induction, demonstrating reversal in rodent models without broad . HSCs exhibit sophisticated mechanical sensing through , which detect (ECM) stiffness and transduce signals promoting activation and fibrogenesis. Increased ECM rigidity, as seen in fibrotic livers, engages β1 and αv pathways to activate YAP/TAZ transcription factors, driving HSC contractility and production. Conceptual biomechanical models illustrate how stiffness gradients (from ~0.5 kPa in healthy liver to >10 kPa in ) reinforce a feed-forward loop, where HSC-derived matrix stiffens the microenvironment further, perpetuating disease. This mechanotransduction is a promising target, with inhibitors like cilengitide showing potential to attenuate HSC responses in advanced .

References

  1. [1]
    Hepatic Stellate Cells: Protean, Multifunctional, and Enigmatic Cells ...
    Stellate cells can be viewed as the nexus in a complex sinusoidal milieu that requires tightly regulated autocrine and paracrine cross-talk, rapid responses to ...
  2. [2]
  3. [3]
    The evidence-based multifaceted roles of hepatic stellate cells in ...
    May 1, 2024 · Hepatic stellate cells (HSCs) play central roles in liver disease pathogenesis, spanning steatosis to cirrhosis and hepatocellular carcinoma.
  4. [4]
    Hepatic Stellate Cells in Physiology and Pathology - PMC
    Hepatic stellate cells (HSCs) are resident mesenchymal cells located in the perisinusoidal area between endothelial cells and hepatocytes known as the Space of ...
  5. [5]
    Roles of hepatic stellate cells in liver inflammation: a new perspective
    Apr 25, 2016 · HSCs reside in the space of Disse, the abluminal side of the sinusoids between liver sinusoidal endothelium and hepatocytes. They represent 5–8 ...<|control11|><|separator|>
  6. [6]
    Intralobular Distribution of Vitamin A-Storing Lipid Droplets in ...
    Jan 14, 2004 · Hepatic stellate cells (vitamin A-storing cells, fat-storing cells, lipocytes, interstitial cells) are located in the perisinusoidal space of ...
  7. [7]
    Hepatic stellate cells: Three-dimensional structure, localization ...
    Hepatic stellate cells (HSCs) are vitamin-A storing, collagen-producing cells in hepatic lobules, forming a 'stellon' with sinusoids and hepatocytes.
  8. [8]
    Hepatic stellate cells – from past till present: morphology, human ...
    Vinculin and cellular retinol-binding protein-1 are markers for quiescent and activated hepatic stellate cells in formalin-fixed paraffin embedded human liver.Missing: reelin | Show results with:reelin
  9. [9]
    a novel marker for the differentiation of HSC from other liver ...
    ... Expression of reelin in hepatic stellate cells and during hepatic tissue repair: a novel marker for the differentiation of HSC from other liver myofibroblasts.
  10. [10]
    Accumulation of Vitamin A in the Hepatic Stellate Cell of Arctic Top ...
    Aug 21, 2012 · In physiological conditions, hepatic stellate cells (HSCs; also called as vitamin ... 50–80% of the total vitamin A in the whole body as ...
  11. [11]
    HEPATIC STELLATE CELL LIPID DROPLETS - PubMed Central - NIH
    All-trans-retinol, which by definition is vitamin A, is routinely referred to simply as retinol. Retinyl esters are retinoid storage forms. Outside of the eye, ...
  12. [12]
    Hepatic stellate cells retain the capacity to synthesize retinyl esters ...
    The liver has the largest capacity to store retinol as retinyl esters. Approximately 70–90% of the retinyl esters are stored in lipid droplets of hepatic ...
  13. [13]
    Metabolic Hallmarks of Hepatic Stellate Cells in Liver Fibrosis - MDPI
    Their main functions are the secretion of laminin, proteoglycans, and type IV collagen to form basement membrane-like structures. Quiescent HSCs start to ...2. Hepatic Stellate Cells · 3.1. Retinol Metabolism · 3.2. Lipid Metabolism
  14. [14]
    Matrix metalloproteinases, the pros and cons, in liver fibrosis - NIH
    Hepatic stellate cells are the principal cell types to produce ECM in normal liver., Isolated HSC undergo spontaneous activation when plated on plastic. Such in ...
  15. [15]
    Hepatic stellate cells require a stiff environment for myofibroblastic ...
    HSC cultured on a basement membrane-like matrix (Matrigel) remain quiescent, and HSC myofibroblasts placed on Matrigel lose their myofibroblastic ...
  16. [16]
    Hepatic stellate cells control liver zonation, size and functions via R ...
    Mar 12, 2025 · We identify R-spondin 3 (RSPO3), an HSC-enriched modulator of WNT signalling, as responsible for these hepatocyte-regulatory effects of HSCs.
  17. [17]
    Hepatic stellate cells regulate multiple aspects of hepatocyte ...
    Jul 7, 2025 · Whilst Wnt signalling is well known to regulate zonation of the liver and RSPO3 expressed by pericentral endothelial cells has previously been ...
  18. [18]
    Hepatic stellate cells promote immunotolerance following orthotopic ...
    Moreover, HSCs stimulated IL-10 and TGF-β production in the tolerance group. This study suggests that HSCs are involved in liver transplantation immune ...
  19. [19]
    Hepatic immune tolerance induced by hepatic stellate cells - PMC
    Hepatic stellate cells (HpSCs) possess weak antigen-presenting ability and function as immunological bystander cells in the regulation of the immune response by ...Missing: NKT | Show results with:NKT
  20. [20]
    Mechanisms of hepatic stellate cell activation - Nature
    May 10, 2017 · Autophagy drives HSC activation by providing energy substrates and is linked to increased endoplasmic reticulum (ER) stress. Oxidative stress is ...
  21. [21]
    Mechanisms of hepatic stellate cell activation - PubMed
    Activation of hepatic stellate cells (HSCs) - transdifferentiation of quiescent, vitamin-A-storing cells into proliferative, fibrogenic myofibroblasts.
  22. [22]
    Extra- and Intra-Cellular Mechanisms of Hepatic Stellate Cell ... - NIH
    In the current review, we discuss pathways of HSC activation, emphasizing emerging extra- and intra-cellular signals that drive this important cellular ...
  23. [23]
    Pathophysiological mechanisms of hepatic stellate cells activation in ...
    The composition of ECM shifts from type IV collagen, heparan sulfate proteoglycan and laminin to type I and III fibrillar collagen. ... collagen in these cells ...
  24. [24]
    Liver fibrosis and hepatic stellate cells: Etiology, pathological ... - NIH
    Importantly, hepatic stellate cells (HSCs) play a key role in the initiation, progression, and regression of liver fibrosis by secreting fibrogenic factors ...
  25. [25]
    NKG2D ligands mediate immunosurveillance of senescent cells - PMC
    In liver fibrosis, the accumulation of senescent activated stellate cells is increased in mice lacking NKG2D receptor leading to increased fibrosis. Overall, ...
  26. [26]
    Friend or foe? The elusive role of hepatic stellate cells in liver cancer
    Aug 7, 2023 · In this Review, we discuss the dual roles of HSC subpopulations in liver fibrogenesis and their contribution to liver cancer promotion, progression and ...
  27. [27]
    Hepatic Stellate Cell Regulation of Liver Regeneration and Repair
    Nov 13, 2020 · Here we discuss how the use of preclinical models are helping to shape our understanding of the role of the mesenchyme during liver regeneration.
  28. [28]
    Hepatic Stellate Cells: Dictating Outcome in Nonalcoholic Fatty Liver ...
    A key mechanism driving fibrosis development during NASH is the activation of hepatic stellate cells; once activated they differentiate into highly ...
  29. [29]
    Insights into the impact of hepatitis B virus on hepatic stellate cell ...
    Apr 11, 2023 · The activation of hepatic stellate cells (HSCs) is a central event in the occurrence and progression of liver fibrosis.
  30. [30]
    Karl Wilhelm Kupffer And His Contributions To Modern Hepatology
    The hepatic stellate cells and reticular fibers are frequently stained concomitantly in gold preparations. Kupffer (1876) emphasized that stellate cells were ...
  31. [31]
    Hepatic Stellate Cells - SpringerLink
    In 1876, von Kupffer discovered star-shaped cells in the hepatic lobules using the gold chloride method, and called them “Sternzellen” (stellate cells).
  32. [32]
    History, heterogeneity, developmental biology, and functions of ...
    In 1971, Wake demonstrated that the Sternzellen of von Kupffer and the fat-storing cells described by Ito were identical. Wake also established that these cells ...Missing: Keith | Show results with:Keith
  33. [33]
    Single Cell, Single Nucleus and Spatial RNA Sequencing of the ...
    Jun 27, 2025 · Single Cell, Single Nucleus and Spatial RNA Sequencing of the Human Liver Identifies Hepatic Stellate Cell and Cholangiocyte Heterogeneity.
  34. [34]
    Hepatic stellate cell heterogeneity: Functional aspects ... - Hepatology
    May 8, 2025 · Hepatic stellate cells (HSCs) represent a highly heterogeneous mesenchymal cell population with multifaceted roles in liver homeostasis and disease.
  35. [35]
    Hepatic stellate cell single cell atlas reveals a highly similar ...
    This study establishes a single-cell atlas of hepatic stellate cells across various liver injuries, highlighting a conserved activation process between ...
  36. [36]
    [PDF] Hepatic stellate cells as RSPO3-Wnt/β - ProBiologists
    May 14, 2025 · Hepatic stellate cells (HSCs) act as hubs for RSPO3-Wnt/β-catenin signaling, regulating liver zonation and regeneration, and are both ...
  37. [37]
    Article Intercellular communication between hepatic stellate cells ...
    Jul 18, 2025 · Hepatic stellate cells (HSCs) play a central role in the development of liver fibrosis. We previously showed that fibroblast growth factor ...
  38. [38]
    HEPATIC STELLATE CELL-DERIVED EXOSOMES MODULATE ...
    In this study we report EXOs derived from activated-HSCs stimulates pro-fibrotic cytokine release in macrophages and increase macrophage migration.
  39. [39]
    [PDF] Exploring hepatic stellate cell-driven fibrosis: therapeutic advances ...
    Aug 4, 2025 · Approach: This review highlights the patho- physiological features of liver fibrosis, with a focus on novel targets in hepatic stellate cells ( ...
  40. [40]
    Targeting Hepatic Stellate Cells for the Prevention and Treatment of ...
    HSC-targeted approaches using specific surface markers and receptors may enable the selective delivery of drugs, oligonucleotides, and therapeutic peptides.Missing: gliotoxin | Show results with:gliotoxin
  41. [41]
    Targeting Hepatic Stellate Cells for the Treatment of Liver Fibrosis ...
    This review serves to summarize the current findings of herbal medicines combining with modern medicines in the area of fibrosis.
  42. [42]
    Mechanical homeostasis imbalance in hepatic stellate cells ...
    Apr 20, 2023 · Type IV collagen, which together with laminin constitutes the major component of the basement membrane of epithelial and endothelial cells ...
  43. [43]
    Increased matrix stiffness promotes fibrogenesis of hepatic stellate ...
    Jun 12, 2025 · We show that cells cultured on a stiff matrix display more accessible chromatin sites, which consist of primed chromatin regions that become more accessible.Missing: sensing | Show results with:sensing
  44. [44]
    Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis ...
    This review explores the primary mechanotransductive signaling networks that drive liver fibrosis, emphasizing how mechanical cues such as increased tissue ...