Published online Nov 26, 2025. doi: 10.4252/wjsc.v17.i11.111090
Revised: July 31, 2025
Accepted: October 9, 2025
Published online: November 26, 2025
Processing time: 156 Days and 16.2 Hours
Hepatic fibrosis is a pathological process characterized by an imbalance between the deposition and degradation of extracellular matrix components. This process is initiated by chronic liver injuries resulting from viral infections, alcoholic liver disease, non-alcoholic fatty liver disease, and autoimmune-mediated hepatic damage. If left untreated, hepatic fibrosis can progress to life-threatening con
Core Tip: Liver fibrosis and cirrhosis are global health challenges resulting from persistent liver injury, hepatocyte damage, inflammation, and hepatic stellate cell activation. These processes dysregulate key signaling pathways such as transforming growth factor beta/small mother against decapentaplegic, Wnt/β-catenin, nuclear factor-kappa B, and mitogen-activated protein kinase, leading to excessive extracellular matrix deposition. Importantly, once the underlying cause is addressed, liver’s natural ability to regenerate and regress fibrosis is restored. Innovative approaches such as mesenchymal stem cell-derived extracellular vesicles and microRNA-based therapies are gaining attention due to their potential to deliver targeted anti-fibrotic molecules, regulate gene expression, and reverse liver fibrosis. Ultimately, these strategies could lessen the burden of chronic liver diseases globally.
- Citation: Islam MA, Mawya J, Salma U, Abu Kasim NH, Haque N. Unravelling the reversion mechanisms of activated hepatic stellate cell properties by extracellular vesicles from mesenchymal stem cells. World J Stem Cells 2025; 17(11): 111090
- URL: https://www.wjgnet.com/1948-0210/full/v17/i11/111090.htm
- DOI: https://dx.doi.org/10.4252/wjsc.v17.i11.111090
The liver is involved in performing crucial functions such as detoxification, metabolic regulation, and immunological support[1,2]. However, chronic liver diseases, particularly cirrhosis and hepatic fibrosis, are major global health concerns. According to the Centers for Disease Control and Prevention, in the United States, liver diseases cause around 40000 deaths annually[3]. Prevalent causes encompass viral hepatitis (B and C), alcoholic liver disease, and non-alcoholic fatty liver disease, the latter impacting up to 25% of Americans and frequently progressing to non-alcoholic steatohepatitis (NASH) and fibrosis[4].
High rates of morbidity and mortality are experienced by patients with chronic liver diseases who develop liver fibrosis and cirrhosis, which significantly burdens society financially[5]. It affects over 800 million people annually and has a mortality rate of about 2 million deaths equivalent to 4% of global mortality worldwide[6]. Cirrhosis-related problems account for one million deaths, whereas viral hepatitis and hepatocellular cancer contribute to the remaining one million deaths[7,8].
Liver fibrosis is a slow-developing, often clinically unmanifested condition where continuous liver injury leads to stiffening and loss of function, potentially progressing to cirrhosis or liver cancer[9-12]. It can be caused by various factors including alcohol, metabolic issues, viral infections, toxins, autoimmune diseases, and prolonged use of certain medications[11,13-15]. A key factor in the pathophysiology of liver fibrosis is the activation of hepatic stellate cells (HSCs). Usually quiescent HSCs (qHSCs) activate in response to liver damage, transforming into myofibroblast-like cells that overproduce extracellular matrix (ECM), leading to scar tissue formation[16]. This activation is driven by signals [like reactive oxygen species (ROS)] from damaged liver cells[17-20] and inflammatory cytokines from immune cells[21,22].
At present, liver transplantation remains the most proven effective intervention for managing advanced liver diseases[23-26]. The understanding and treatment of fibrosis are undergoing a significant shift. Previously, fibrosis was considered irreversible with no specific treatments. However, new research is overturning these long-held beliefs. A prime example of this evolving therapeutic landscape is the 2024 Food and Drug Administration approval of resmetirom, the first drug specifically developed for liver fibrosis and NASH. This approval highlights a major change in how fibrosis is viewed and managed therapeutically[27-29]. The potential of phytochemicals and other drugs in partially reversing the condition of fibrosis have been identified by a number of etiological and preclinical studies[30-35]. Although many mediators and routes implicated in hepatic fibrogenesis have been identified, a clear therapeutic drug to treat liver fibrosis is still elusive. Current therapeutic drugs frequently have poor selectivity, limited solubility, and low liver accumulation, which can result in non-target organ absorption and insufficient therapeutic doses in vivo. For example, poor water solubility, low absorption, and imprecise targeting hinder the useful chemicals curcumin and silymarin[36-38].
Recent studies have shown that extracellular vesicles (EVs) produced from mesenchymal stem cells (MSCs) have the potential to control HSC activity, reducing liver fibrosis and promoting tissue healing. For instance, MSC-derived EVs have been shown to inhibit the Wnt/β-catenin signaling cascade in HSCs, resulting in reduced activation and fibrosis in rat models of liver injury[39]. Furthermore, it has been revealed that EVs produced from human liver stem cells reduce the activated phenotype of HSCs; miR-146a-5p is a key mediator of this anti-fibrotic impact[40]. MSC-derived EVs have also been shown to reduce the expression of pro-fibrotic markers in liver spheroids and activated HSCs (aHSCs) that are driven by transforming growth factor beta 1 (TGF-β1), highlighting their potential as a therapy strategy for hepatic fibrosis[40,41]. Hence, this review will concentrate on the activation of HSCs, pathogenesis, and mechanisms of liver fibrosis, along with its clinical implications, by examining the cellular and molecular pathways of liver fibrosis resolution and the reversion mechanisms of aHSCs using EVs from MSCs.
The liver maintains a perfect balance between building up and breaking down its ECM, a vital support structure. The ECM, a complex network of macromolecules, regulates numerous physiological processes via various signaling pathways. These processes include cell division, adhesion, migration, proliferation, growth, metabolism, injury healing, and tissue remodeling[42,43]. Chronic liver disease disrupts equilibrium, leading to more production than breakdown[44]. Liver fibrosis is characterized by continuous ECM deposition[45], harming the liver’s structure[5] and impairing normal activity[12]. Active interaction between immune, non-parenchymal, and hepatocytes causes the intricate process[35,46,47].
ROS, generated by various stimuli such as toxins, viruses, cholestasis, hypoxia, and insulin resistance, contribute to hepatocyte damage, apoptosis, steatosis, and infiltration of immune cells, especially Kupffer cells (KCs)[48,49]. Simultaneously, sinusoidal endothelial cells undergo capillarization, characterized by the loss of fenestrae, which alters sinusoidal structure[50]. This process is initiated by chronic hepatocyte injury, leading to the release of pro-fibrotic cytokines and growth factors-including tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, TGF-β, and platelet-derived growth factor (PDGF) through interactions with natural killer cells and KCs. Primary responder cells, such as HSCs, become activated, resulting in excessive ECM deposition[51,52]. Although HSCs are the central mediators in liver fibrosis, interactions among various liver cell types including hepatocytes, liver sinusoidal endothelial cells (LSECs), and inflammatory cells are also involved in the fibrotic process[10]. The process of liver fibrosis, and the main functions of different cells and cytokines is briefly described below.
The liver consists of parenchymal cells and non-parenchymal cells. Parenchymal cells, account for 80% of the liver’s volume and 60% of its total cell count[10]. Hepatocytes are crucial for detoxification, energy metabolism, and lipid, protein, and bile secretion[53]. Because of its nature, liver to be affected by the xenobiotic drugs and toxins, which lead to the hepatocyte death. Dead hepatocytes release damage-associated molecular patterns (DAMPs), which encircle KCs and HSCs, leading to inflammation and fibrosis[54,55].
One key DAMP, high-mobility group box-1 (HMGB1), can directly activate HSCs, especially in conditions like hepatitis B[56]. Besides, damaged hepatocytes can also release exosomes carrying signaling molecules and microRNAs (miRNAs) particularly miR-27a that could further boost HSC activation, suppress aHSC autophagy and eventually could accelerate liver scarring in metabolic-associated fatty liver disease[57]. In brief, healthy hepatocytes keep the liver running smoothly, but their damage and death are critical triggers in the complex process of liver inflammation and scarring.
KCs, the liver’s resident macrophages, are a vital part of the innate immune system and are essential for maintaining liver homeostasis[58,59]. KCs exhibit a dual role in liver fibrosis[60]. During fibrosis progression, KCs exacerbate fibrosis by recruiting more inflammatory cells to the injured liver, increasing the production of pro-inflammatory cytokines and chemokines, and activating HSCs to produce excessive fibrotic tissue. In contrast, during fibrosis regression, macrophages change their function, aiding recovery by producing matrix-degrading enzymes like matrix metalloproteinase (MMP)-9 and MMP-2 to dismantle ECM. This resolution phase also entails lowering the production of inflammatory and fibrogenic mediators[61].
In case of persistent liver injury inflammation is triggered. Damaged cells release ROS and other signaling molecules activate immune cells, specifically lymphocytes and macrophages[62]. As a consequence, activated KCs rapidly secrete a variety of inflammatory chemicals, namely IL-1β, TNF-α, chemokine (C-C motif) ligand 2, and chemokine (C-C motif) ligand 5[5,10,63]. Furthermore, KCs activation can increase nuclear factor kappa-B (NF-κB) in HSCs, which further promotes the release of pro-inflammatory cytokines. The release of these substances worsens liver fibrosis and directly activates HSCs, the primary collagen-producing cells[64].
Non-parenchymal cells called LSECs make up the sinusoidal wall and serve as the contact between hepatocytes and HSCs on one side and the blood and KCs on the other. In a healthy liver, LSECs are considered “guardians of hepatic homeostasis” due to their vasodilatory, anti-inflammatory, antithrombotic, anti-angiogenic, antifibrotic, and regeneration-promoting properties[65]. HSCs are kept in a resting condition by endothelium-derived nitric oxide, which is mostly produced by LSECs. When liver damage occurs, LSECs become capillarized, which can increase the synthesis of vasoconstrictors (endothelin 1, thromboxane A2, and angiotensin II) and decrease the production of vasodilators (like nitric oxide, cyclooxygenase, and prostaglandin I2). This imbalance causes inflammation and liver fibrosis in addition to changing the LSEC phenotype and activating HSCs[66-68]. Additionally, LSECs can activate signaling pathways like Wnt/β-catenin or release signaling molecules like TGF-β and PDGF, which in turn stimulate HSCs[35].
HSCs are found in the liver’s subendothelial region. In a healthy liver, they remain quiescent. Their normal functions include storing fat and metabolizing vitamin A, working closely with surrounding hepatocytes and sinusoidal endothelial cells[69,70]. qHSCs play a crucial role in maintaining the ECM homeostasis by secreting moderate amounts of ECM proteins like collagen type III, collagen type IV, and laminin[35,71]. They also produce MMPs, such as MMP-1, which help break down ECM. Tissue inhibitors of MMP (TIMP)-1 and TIMP-2, two TIMPs, are also released by qHSCs. TIMP-1 is particularly important as it inhibits MMPs and helps prevent ECM breakdown, ensuring a healthy balance between ECM production and degradation[72-75]. This precise interplay between MMPs and TIMPs is vital for liver matrix regeneration and maintaining healthy liver architecture[76].
One crucial stage in hepatic fibrogenesis is the activation of qHSCs[77]. This activation is triggered by various factors, such as inflammatory stimuli, fibrogenic cytokines like TGF-β, ROS, activated KCs, platelets, and products from injured hepatocytes. Upon activation, aHSCs not only secrete large amounts of ECM (primarily collagen I and III) but also release cytokines like TGF-β that sustain their own activation, creating a vicious cycle[78]. Besides, a significant increase in the expression of TIMP-1, further hindering ECM breakdown[78]. This continuous activity leads to the excessive deposition of mature collagen fibers in the Disse space, causing liver fibrosis[79].
Among several pathways that are involved in the liver fibrosis TGF-β/small mother against decapentaplegic (SMAD), Wnt/β-catenin, NF-κB pathway, mitogen-activated protein kinase (MAPK) pathways are considered the most important. The roles of these pathways are described in brief below.
The TGF-β/SMAD pathway plays a central and critical role in the development of liver fibrosis and cirrhosis, primarily through its influence on HSC activation and ECM production. TGF-β is a potent fibrogenic cytokine released by injured hepatocytes, inflammatory cells, and KCs during liver injury. TGF-β binds to TGF-β receptors (type I and II) on HSCs, initiating intracellular signaling cascades. Upon TGF-β binding, type I TGF-β receptor phosphorylates receptor-regulated SMADs (SMAD2 and SMAD3). Phosphorylated SMAD2/3 forms a complex with SMAD4, then translocate into the nucleus. Inside the nucleus, the SMAD complex regulates the transcription of target genes involved in fibrosis[80,81]. Upregulated genes encode ECM components, especially collagen types I and III. Stimulates alpha-smooth muscle actin (α-SMA) expression, marking the transdifferentiation of qHSCs into aHSC[82]. Enhances production of TIMPs that eventually reduce ECM degradation. Elevated TGF-β levels sustain HSC activation and fibrogenesis[83].
The Wnt/β-catenin signaling pathway plays an important role in liver fibrogenesis and the activation of HSCs during the progression to cirrhosis. Glycoproteins called Wnt proteins are secreted and bind to cell surface Frizzled receptors and co-receptors (like lipoprotein receptor-related protein 5/6. In the absence of Wnt signaling, a destruction complex that contains glycogen synthase kinase-3β and other elements targets β-catenin for degradation. When Wnt binds to its receptors (such as Frizzled and lipoprotein receptor-related protein 5/6), this destruction complex is inhibited, preventing β-catenin degradation. As a result, β-catenin stabilizes and accumulates in the cytoplasm. The accumulated β-catenin then translocates into the nucleus, where it interacts with T-cell factor/Lymphoid enhancer factor transcription factors to regulate the expression of target genes. These genes are involved in critical cellular processes such as proliferation, survival, and differentiation[84,85].
The NF-κB signaling pathway is reported in controlling inflammation, fibrosis, and even the development of hepatocellular carcinoma[86]. NF-κB primarily influences liver scarring through three main cell types: (1) By regulating hepatocyte damage, which acts as the primary trigger for fibrogenic responses; (2) By controlling inflammatory signals generated by macrophages and other immune cells within the liver; and (3) By affecting the fibrogenic activity of HSCs. NF-κB is expressed in nearly all cell types and controls a wide array of genes involved in innate and adaptive immunity, cell growth, differentiation, survival, and apoptosis[87]. Two main pathways activate NF-κB: The canonical pathway involving RELA (p65) and the non-canonical pathway involving RELB. Activation of NF-κB results in the transcription of hundreds of genes containing κB binding sites, most of which regulate inflammation, immune responses, and cell survival[88]. Abnormal or excessive NF-κB activation is observed in many chronic liver diseases, including alcoholic steatohepatitis, NASH, and viral infections, highlighting its vital role in maintaining liver homeostasis, immune cell interactions, and tissue repair processes. More specifically, NF-κB signaling upregulates the expression of activation markers, such as α-SMA and fibrogenic genes like collagen types I and III. NF-κB drives the expression of pro-inflammatory cytokines (e.g., IL-6, monocyte chemotactic protein-1) and chemokines, which recruit immune cells to the liver, sustain inflammatory milieu and further promote HSC activation and fibrosis progression[21,22]. NF-κB promotes HSC survival by inducing anti-apoptotic genes, thereby prolonging their activated state. This contributes to persistent fibrogenic activity in chronic liver injury.
The MAPK pathway influences processes like inflammation, cell death, and fibrosis, with specific MAPK members like p38 and c-Jun N-terminal kinase (JNK) implicated in the progression of liver injury and fibrosis[89]. MAPK cascades are signaling pathways that carry external inputs to the nucleus. Each of the three primary MAPK cascades-which have been thoroughly described-converges on extracellular signal-regulated kinases (ERKs), JNKs, and p38 MAPKs and is made up of three classes of serine/threonine kinases: MPKKkinase, MAPK, and MAPK kinase[90,91]. p38 MAPK is activated during liver injury and participates in inflammatory responses, potentially contributing to liver damage and fibrosis. Inhibition of p38 MAPK may be beneficial in treating liver cirrhosis. JNK consists of three isoforms-JNK1, JNK2, and JNK3-encoded by distinct genes. JNK1 and JNK2 are expressed in nearly all tissues, including the liver, while JNK3 is primarily found in neuronal tissue[92]. JNK plays versatile roles in regulating cellular processes such as proliferation, differentiation, and apoptosis, primarily through phosphorylation and modification of downstream targets. Its activity is stimulus- and cell-type dependent. ERK1/2 are part of the MAPK pathway that governs cell proliferation, differentiation, and development. They are activated by signaling[93]. In the liver, ERK1/2 promotes autophagy in hepatocytes, which can help reduce liver steatosis.
The Notch signaling pathway is highly conserved and plays essential roles in liver development, biliary cell differentiation, and fibrosis[94]. In fibrotic liver tissue, abnormal activation of Notch encourages the ductular reaction and the proliferation of reactive cholangiocytes, which release pro-fibrotic cytokines like TGF-β1 and PDGF that, in turn, activate HSCs[95,96]. Moreover, Notch promotes epithelial-to-mesenchymal transition-like changes in cholangiocytes and increases the expression of fibrogenic genes such as α-SMA and collagen type I. Experimental studies have shown that blocking Notch signaling with γ-secretase inhibitors can reduce fibrosis, highlighting its potential as a therapeutic target[97,98].
The Hedgehog signaling pathway remains inactive in the healthy adult liver but becomes reactivated in response to liver injury. When Hedgehog ligands-Sonic Hedgehog, Indian Hedgehog, or Desert Hedgehog-bind to the Patched receptor, they relieve the suppression of smoothened, leading to activation of GLI transcription factors (GLI1-3). This activation promotes the survival, proliferation, and activation of HSCs, and also attracts immune cells such as macrophages and natural killer T cells, which contribute to inflammation and fibrosis. Hedgehog signaling supports the maintenance of myofibroblast-like cells and stimulates the expression of pro-fibrogenic genes, resulting in increased ECM production. In models of cholestasis and NASH, activation of the Hedgehog pathway has been directly linked to the severity of fibrosis. Pharmacological inhibition of this pathway has been shown to reduce collagen accumulation and attenuate HSC activation in vivo[99-101].
The Hippo signaling pathway is essential for essential for controlling tissue regeneration, cell division, and organ growth[102]. The downstream effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are dephosphorylated and go into the nucleus when Hippo signaling is interfered with. There, they interact with YAP-transcriptional enhancer factor domain family member transcription factors. Genes like connective tissue growth factor, CYR61, and amphiregulin that are involved in cell development and fibrogenesis are expressed more frequently as a result of this interaction. YAP is elevated in hepatocytes, cholangiocytes, and HSCs in chronic liver damage, which causes enhanced proliferation and a fibrogenic phenotype. The crucial role of YAP in liver fibrogenesis is highlighted by the fact that blocking YAP activity significantly lowers ECM deposition, while experimentally activating YAP in hepatocytes or cholangiocytes causes rapid fibrosis in murine models[103-106].
Liver fibrosis is orchestrated by a complex interplay among several signaling pathways that regulate HSC activation, inflammation, and ECM remodeling. These pathways include the TGF-β/SMAD axis, Wnt/β-catenin signaling, the NF-κB inflammatory cascade, MAPK signaling modules, and PDGF-mediated proliferative signals. Importantly, these pathways exhibit extensive cross-talk, creating amplifying feedback loops that sustain fibrogenesis[5,60,83,107-113].
TGF-β/SMAD signaling, a central driver of fibrosis, not only induces ECM components such as type I and III collagen and α-SMA via SMAD2/3 activation, but also interfaces with other pathways[114-116]. Beyond its direct effects, TGF-β interacts with other signaling pathways to amplify fibrogenic responses. It enhances Wnt/β-catenin signaling by downregulating antagonists like Dickkopf-1, leading to stabilization and accumulation of β-catenin. The stabilized β-catenin then cooperates with SMAD proteins at promoters of pro-fibrotic genes, further increasing their transcription[117-119].
Additionally, TGF-β can indirectly activate NF-κB signaling via TGF-β-activated kinase 1, resulting in the upregulation of pro-fibrotic cytokines such as IL-6 and monocyte chemotactic protein-1, as well as increased TGF-β expression itself[120]. Pro-inflammatory cytokines like TNF-α and IL-1β, released by KCs and damaged hepatocytes, stimulate NF-κB and MAPK pathways (including ERK and JNK), which in turn modulate transcription factors to enhance SMAD signaling and promote fibrogenesis[121]. Furthermore, NF-κB and TGF-β increase TIMP expression, limiting MMP-mediated ECM degradation and promoting net ECM accumulation[122,123]. This intricate network underscores the interconnected nature of fibrogenic signaling pathways in liver injury[121].
The MAPK pathway plays a significant role in liver fibrosis through both direct and indirect mechanisms. JNK, a member of the MAPK family, can phosphorylate SMAD3 at linker regions, altering its DNA-binding affinity and transcriptional activity, thereby influencing fibrogenic gene expression. Meanwhile, ERK promotes proliferation and survival of aHSC[124,125]. Additionally, MAPKs are activated by PDGF, a potent mitogen for HSCs, which works synergistically with TGF-β to sustain aHSCs proliferation and fibrogenesis[126]. Furthermore, DAMPs, such as HMGB1, initiate fibrogenic signaling by binding to pattern recognition receptors like Toll-like receptor 4. This engagement leads to rapid activation of NF-κB and MAPK pathways, which amplify cytokine production and reinforce the fibrotic response, creating a feedback loop that accelerates liver fibrosis progression[127].
The Notch, Hippo, and Hedgehog pathways form a critical regulatory axis in fibrogenesis, particularly influencing biliary proliferation, ductular reaction, and stromal activation[128-130]. The transcriptional co-activators YAP/TAZ, effectors of the Hippo pathway, promote expression of Notch ligands (e.g., Jagged1), enhancing Notch signaling in cholangiocytes and aHSCs. Conversely, Notch activation supports YAP nuclear localization, reinforcing profibrotic transcriptional programs. Additionally, YAP/TAZ interact with GLI transcription factors of the Hedgehog pathway, while Hedgehog signaling reciprocally upregulates YAP expression. This reciprocal feed-forward loop drives HSC activation, progenitor cell expansion, and ECM production[95,130-134].
ECM remodeling is controlled by the balance between MMPs and TIMPs. TGF-β, Notch, and YAP/TAZ signaling promote TIMP1/2 expression, limiting ECM degradation and favoring matrix accumulation. In contrast, early activation of MAPKs and inflammatory cytokines transiently promotes MMP expression to facilitate cell migration. This dynamic regulation ensures both tissue remodeling and matrix deposition are coordinated during fibrogenesis[135,136].
Pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α, along with DAMPs like HMGB1, serve as upstream initiators of fibrosis by activating Toll-like receptor, NF-κB, Janus kinase (JAK)/signal transducer and activator of the transcription, and MAPK pathways. These signals stimulate the release of TGF-β, PDGF, and Wnt ligands, indirectly modulating Hippo, Notch, and Hedgehog activity[120,121,137]. Additionally, ROS generated during chronic injury further enhance TGF-β expression and ECM gene transcription, establishing a vicious cycle of injury and fibrogenesis[138-140]. These synergistic and feedback interactions create a robust, self-perpetuating fibrotic program. Therefore, therapeutic strategies that target multiple key nodes within this integrated network rather than isolated pathways may prove more effective in halting or reversing liver fibrosis associated with chronic liver diseases.
Cell-based therapies, particularly those involving MSCs, have emerged as promising alternatives for treating chronic liver diseases. MSCs are multipotent stem cells known for their tissue repair and immune regulation capabilities. They exert anti-fibrotic effects through various mechanisms, including modulating hepatic immune responses, secreting trophic cytokines to reduce hepatocyte apoptosis, exhibiting antioxidant properties, inhibiting HSC proliferation, and regulating TIMP-1 expression[89,141]. MSCs can be sourced from various tissues, such as bone marrow, liver, and umbilical cord. While MSC transplantation shows promise in improving liver function in conditions like alcoholic cirrhosis, a potential limitation is their capacity to differentiate into myofibroblasts, which could inadvertently contribute to fibrosis[142-145].
Recently, attention has shifted to EVs, particularly exosomes derived from MSCs, as a novel therapeutic strategy for liver fibrosis. miRNAs are small non-coding RNAs that regulate key cellular processes such as division, differentiation, and cell death. Over 1000 human miRNAs have been identified, with estimates suggesting they control about one-third of gene expression[146]. The human genome comprises approximately thirty collagen genes that generate different types of collagen fibrils vital for cellular functions. Clinical trials are being conducted to test the safety and effectiveness of MSC-EVs for a variety of disorders[147]. MSC-derived exosomes carry a similar therapeutic payload as their parent MSCs and can significantly reduce HSC activation[148,149]. They offer advantages over MSCs, such as smaller size, reduced complexity, lower immunogenicity, and no risk of tumor development. These characteristics make exosomes promising natural nanovectors for therapeutic agents in hepatic fibrosis treatment[150,151].
A key mechanism by which MSCs and their EVs exert their anti-fibrotic effects is through the transfer of miRNAs[146]. They play a crucial role in regulating gene expression in nearly all cellular functions and are implicated in various liver conditions, including fibrosis[40]. During liver fibrosis, miRNAs can be either up- or down-regulated. Specific miRNAs, such as miR-125b from MSC-derived exosomes, can prevent HSC activation by suppressing smoothened expression[152]. Similarly, miR-122 from adipose tissue-derived MSCs can inhibit HSC activation and proliferation[153,154].
Furthermore, miRNAs are involved in regulating collagen accumulation and the TGF-β/SMAD signaling pathway, both central to liver fibrosis. miRNAs like let-7 g, miR-29b, and miR-29c directly target collagen expression, while miR-29a suppresses collagen type 1 alpha 1 (COL1A1) expression and promotes apoptosis[155-157]. miRNAs such as miR-30e, miR-133b, and miR-19b have been shown to control collagen production and inhibit fibrotic pathways by targeting key components like prolyl 4-hydroxylase 2 (P4HA2) and TGF-β signaling[158-160]. Moreover, a range of miRNAs, including miR-15a, miR-15b, miR-16, miR-29a, miR-125b, miR-126, miR-146a, miR-195, miR-199a, and miR-200, have been identified as having roles in reversing fibrosis by regulating genes involved in HSC activation and the TGF-β pathway[161,162]. miR-21, miR-29a, miR-146a, miR-125b, miR-126, miR-195, and miR-199a are frequently identified in MSC-derived microvesicles across different tissue sources[163,164]. These miRNAs contribute to the regenerative, anti-inflammatory, and pro-angiogenic effects of MSCs and their derived EVs.
The following succinctly describes the potential role of different cells and molecules in reversing the pathogenicity of liver fibrosis. An increasing number of clinical studies indicate that fibrosis can partially regress once the underlying cause is eliminated, challenging earlier beliefs that the condition was irreversible[165-167]. Unlike organs such as the kidney and lung, the liver possesses a greater capacity for regeneration, making the regression of fibrosis and restoration of normal tissue structure more likely. These characteristics may simplify the development of effective therapies for liver fibrosis[9,167].
It is required to find more effective medicines to postpone fibrosis progression or encourage regression of liver fibrosis, which can assist prevent the advancement of cirrhosis or cancer[70,165]. Understanding the mechanisms underlying liver fibrosis regression can help identify new treatment targets. The main ways to achieve liver fibrosis regression[167] are to eliminate the factors causing liver damage, reduce the number of aHSCs[168], inhibit the signal pathways linked to liver fibrosis[169], and promote the degradation of the ECM[170].
The intricate mechanisms underlying liver fibrosis involve changes in cell populations, cytokines, and their molecular pathways[171]. Notably, activation of HSCs is a hallmark feature of liver fibrosis[171,172]. Upon removal of chronic liver injury, fibrosis tends to regress, and aHSCs either decrease in number or disappear[173]. Additionally, both experimental and clinical studies have demonstrated that apoptosis contributes to the elimination of aHSCs[174], or while inactivation into a quiescent-like state[175,176], or senescence[177] may also facilitate fibrosis regression. Due to their pivotal role in fibrosis development, aHSCs are considered a primary target for therapeutic intervention aimed at disease regression[178]. Therefore, strategies that reduce the number of aHSCs such as inhibiting their proliferation or activation[177-179], promoting apoptosis or autophagy[171,172,180], and deactivating the aHSCs[175,176] may serve as effective approaches to reverse liver fibrosis[181].
Apoptosis, a form of programmed cell death, aids in regulating the HSCs’ equilibrium between growth and death[167]. The condition may worsen as a result of HSCs’ abnormal activation and proliferation during liver fibrosis progression, which may hasten the death of other healthy liver cells[54,182]. Clinical studies revealed that, regardless of HSC activation or proliferation, the quantity of apoptotic HSCs declined as the fibrosis stage increased[183]. Research on animal models of liver damage caused by various techniques has demonstrated a strong correlation between the regression of fibrosis and the reduction of HSCs through apoptosis induction, indicating that HSC apoptosis is also a key factor in the regression of liver fibrosis[174,183]. Thus, increasing the rate at which aHSCs die and decreasing the overall quantity of aHSCs can reverse liver fibrosis[184,185]. The researchers have shown that liver fibrosis treatment could rely on endoplasmic reticulum stress (ERS)-mediated apoptosis[186]. Additionally, in the HSC line HSC-T6, the
The liver is the primary site of expression for the metabolic enzyme alcohol dehydrogenase (ADH), and ADH-I is the traditional liver ADH among these family members[187]. According to research, the human fibrotic liver exhibits substantially greater ADHI activity than the normal liver. ADH-I may enhance the proliferation, motility, adhesion, and invasion of HSCs[188]. Therefore, ADH-I might be a target for future hepatic fibrosis medications.
Anti-fibrotic medications target HSCs because of their significant role in liver fibrosis[52]. Focusing on aHSC deactivation has become a novel and effective approach to liver fibrosis treatment[176,189]. Fibrosis experimental models consistently show that fibrosis can reverse when aHSCs are eliminated by apoptosis or other mechanisms[190,191]. Recent research has demonstrated that transcriptional reprogramming, including ectopic expression of GATA-binding protein 4 (GATA4), forkhead transcription factor 3, hepatocyte nuclear factor 1 homeobox A, and hepatocyte nuclear factor 4 homeobox A in vivo, can convert aHSCs into qHSCs[192].
It has been shown that abnormal ECM component development and HSC activation in the embryonic liver occur when the zinc finger transcription factor GATA4 in HSCs is deactivated[189]. Reactivation of GATA4 has been found to reverse liver fibrosis by transforming aHSCs into a qHSCs[82]. Therefore, GATA4 might be a suitable target for liver fibrosis resolution. In another study, the activation of HSCs has been linked to peroxisome proliferator-activated receptor-γ (PPAR-γ)[175]. Significantly lower levels of PPAR-γ are observed during HSC activation[193], and a shift to qHSCs could be brought by PPAR-γ overexpression in aHSCs[194,195]. Transcription factor 21 has been found to be a deactivation factor of aHSCs[196]. Both in vitro and in vivo experiments, overexpression of transcription factor 21 in aHSCs reduced the expression of genes linked to fibrosis, moderately reverse the cells into a quiescent state, and eventually helped to decrease and improved liver function[196].
Excess ECM removal is one of the main goals in treating liver fibrosis. Collagen is the ECM protein that is most common in liver fibrosis. A small interfering RNA to the procollagen α1(I) gene has now been shown to significantly reduces the synthesis of collagen 1 fibrils in animals, and in fibrosis models[197]. This could be another modality to treat liver fibrosis.
The entire process of liver fibrosis involves a number of molecular mechanisms, signaling channels, and cytokines. The disease can be reversed by interfering with these substances and pathways[178,198]. Many signals are crucial to the progression of liver fibrosis, including TGF-β[199], Wnt/β-catenin[200], Notch[128], Hedgehog[201], Hippo[185,202], and inflammasome signaling pathways[203]. From initial liver damage to cirrhosis and fibrosis, TGF-β, the most potent fibrogenic cytokine, influences each stage of the disease’s progression[9,204]. TGF-β is increased during liver fibrosis and primarily acts to stimulate HSCs. Additionally, it can directly stimulate the creation of interstitial fibrillar collagens and increase the synthesis of TIMPs[205]. Consequently, inhibiting TGF-β or interfering with its downstream signaling pathways offers an effective approach to prevent liver fibrosis[206]. Strategies such as targeting specific TGF-β isoforms or blocking TGF-β receptor activation have been explored extensively to disrupt TGF-β-mediated signaling and suppress fibrosis progression[205]. Numerous cytokines, including IL, can activate the JAK signaling pathway, which is crucial to the pathophysiology of hepatic fibrosis. According to studies, using the JAK2 receptor antagonist TG101348 in animal models can lessen hepatic fibrosis[207-209]. The Notch signaling system may be one of the targets for reversing hepatic fibrosis because it can also be used to treat liver fibrosis[10]. Beyond the pathways previously mentioned, liver fibrosis reversal can also be achieved by promoting ECM degradation, boosting immune cell activity and their signaling pathways, employing stem cell transplantation, and applying other innovative approaches. However, further clinical trials are necessary to establish effective methods for reversing liver fibrosis.
A number of cytokines control HSC activation and aHSCs are closely linked to liver fibrosis. miRNAs regulate gene expression post-transcriptionally by binding to target mRNAs, leading to their degradation or translational repression. Given their ability to modulate key pathways involved in liver pathology, miRNAs are closely linked to various liver conditions, including liver fibrosis, making them promising therapeutic targets. During liver fibrosis, the expression levels of specific miRNAs can be both up- and down-regulated. Anti-miRNA oligonucleotides and miRNA masking can reverse up-regulated miRNA[207-209]. In contrast, some studies have observed that downregulated miRNAs may actually play a role in inhibiting liver fibrosis[210-212]. According to reports, miRNAs have antifibrogenic qualities and have the ability to regulate the production of genes including COL1A1, ACTA2, TGF-β receptor type-1, and TIMP1 that are involved in the activation of HSCs[79,176,189]. The role of different miRNAs in regulating pathways involved in liver fibrosis has been extensively studied, revealing their crucial influence on the progression or inhibition of fibrogenic processes. Below are an overview of key miRNAs and their functions.
Many miRNAs influence the production and regulation of collagen, and miR-21 playing major role in controlling the expression of collagen type 1 in particular cell types[213]. miRNAs such as let-7 g, miR-29b, and miR-29c directly target the expression of various collagens by binding to their 3’ untranslated regions[157]. Collagen synthesis induced by TGF-β is activated in HSCs, which play a role in liver fibrosis. Due to its anti-fibrotic effects, miR-29a directly suppresses COL1A1 expression and promotes apoptosis[155,156]. The integrity of the collagen triple helix depends on the hydroxylation of proline residues on collagen, which is mediated by collagen prolyl 4 hydroxylase. Collagen prolyl 4 hydroxylase is an α2β2 tetramer made up of three isoenzymes comprising various catalytic subunits (encoded by the genes P4HA1, P4HA2, and P4HA3) and an a-subunit (the gene P4HB)[214]. P4HA2 promotes collagen deposition in the liver both in vitro and in vivo, which accelerates the development of liver fibrosis and liver cancer. miR-30e, targeting P4HA2 mRNA, controls collagen production[158]. In addition, miR-133b and miR-19b have been shown to inhibit fibrotic pathways by targeting TGF-β signaling components, thereby reducing collagen accumulation[159,160].
In numerous conserved biological signaling cascades, miRNAs play a role in post-transcriptional gene regulation[215,216]. A component of these signaling cascades is the TGF. miRNAs regulate the TGF-β signaling pathway. miRNAs are involved in many biological processes including regulation of apoptosis, development, signal transduction, cell proliferation, and immune defense[217]. Almost every biological activity and cell type, including those in the liver, is targeted and regulated by miRNAs, which also affect gene expression in almost every cellular function. Multiple research studies have demonstrated that alterations in intracellular miRNAs such as miR-155, miR-132, miR-21, miR-26a, and miR-217 are linked to various liver disorders[218-220]. Specifically, miR-155 has been shown to be elevated in KCs following alcohol consumption, with TNF identified as a target of miR-155 that promotes liver inflammation[218]. Additionally, TGF-β1 exerts its biological functions through activation of both SMAD-dependent and SMAD-independent signaling pathways. It is well established that TGF-β primarily mediates its effects by activating downstream mediators SMAD2 and SMAD3, while its activity is negatively regulated by the inhibitory SMAD7[221]. In contrast, miR-15a, miR-15b, miR-16, miR-21, miR-29a, miR-122, miR-125b, miR-126, miR-146a, miR-150-5p, miR-195, miR-199a, miR-200, miR-223, and miR-486-5p have the role in reversing the fibrosis (Table 1). For instance, miR-200 family known to target zinc-finger E-box-binding homeobox 1 and zinc-finger E-box-binding homeobox 2, which are downstream of TGF-β signaling and involved in epithelial-to-mesenchymal transition. They can also directly target components of the TGF-β pathway to inhibit fibrosis[161]. miR-29 family, besides regulating collagen genes, miR-29 can target TGF-β receptor type-2 and SMAD3, reducing TGF-β signaling and fibrotic responses[162].
| miRNA | Target protein/pathway | Mechanism | Ref. |
| miR-29a/b/c | TIMP, SMAD, TGF-β, PDGF, Wnt, MMP, HMGB-1, TLR-4 | Directly targets collagen genes, reduces ECM production, increases MMPs via TIMP suppression, inhibits Wnt signaling | [12,155,156,162] |
| miR-146a | NF-κB, IL-6, TNF-α, IL-1β, MCP-1, TGF-β, Wnt | Targets IRAK1, TRAF6 to inhibit NF-κB pathway; reduces pro-inflammatory cytokines, inhibits HSC activation | [40,222] |
| miR-125b | TGF-β, SMAD, Wnt | Targets SIRT7, suppresses TGF-β/SMAD and Wnt pathways, regulate Hedgehog signaling | [152] |
| miR-122 | Collagen I/III, TIMP, α-SMA | Downregulated in fibrosis; restoration reduces collagen and HSC activation | [153,154] |
| let-7 g | TGF-β, SMAD | Inhibits TGF-β signaling, reduces ECM production, inhibit migration of HCC | [157] |
| miR-30a/e | TGF-β, α-SMA | Targets Beclin-1 and autophagy pathways, suppresses HSC activation | [158] |
| miR-133b | TGF-β | Directly targets TGF-β, inhibits HSC activation | [159] |
| miR-19b | TGFβR2 | May inhibit TGF-β pathway via receptor targeting, inhibit fibrogenesis | [160] |
| miR-26a | Notch, TGF-β | Inhibits HSC proliferation, collagen production, EMT | [223] |
| miR-15a/b, miR-16 | Bcl-2, TGF-β/SMAD, Notch, Wnt | Inhibit HSC proliferation and collagen expression | [224] |
| miR-126 | p5, PI3K-AKT | Maintains endothelial integrity; fibrosis role less defined; regulates HSCs cycle and maintains HSCs | [225] |
| miR-199a/199a-3p | Collagen I/III, caveolin-2, GSK, MAPK | Improve chemo-sensitivity through mTOR pathway | [151,163] |
| miR-200 | EMT, TGF-β, SMAD | Inhibits EMT and TGF-β induced fibrogenesis through SMAD pathway | [161] |
| miR-223 | NF-κB, PDGF, MCP-1, HMGB1 | Inhibits inflammasome activation, reduces inflammation and fibrosis | [226-228] |
| miR-150-5p/miR-150 | TGF-β, c-Myb | Suppresses HSC activation | [229] |
| miR-486-5p | Hedgehog signaling | Suppresses HSC activation and fibrosis | [148] |
Liver fibrosis and cirrhosis represent significant global health challenges characterized by complex cellular and molecular mechanisms, notably the activation of HSCs and dysregulation of key signaling pathways such as TGF-β/SMAD, Wnt/β-catenin, NF-κB, and MAPK. The progression of these conditions results from persistent liver injury, involving hepatocyte damage, inflammatory responses, and alterations in liver cell interactions that promote excessive ECM deposition. Importantly, emerging research highlights the liver’s inherent capacity for regeneration and fibrosis regression once the underlying cause is addressed. Therapeutic strategies targeting HSC activation, promoting apoptosis, inhibiting pro-fibrotic cytokines, and modulating signaling pathways show promise. Innovative approaches such as MSC-derived EVs and miRNA-based therapies are gaining attention due to their potential to deliver targeted anti-fibrotic molecules, regulate gene expression, and reverse fibrosis effectively. While current treatments are limited, these advances suggest a hopeful future for more effective, targeted therapies capable of halting or reversing liver fibrosis, ultimately reducing the burden of chronic liver diseases worldwide.
The authors would like to thank Md Sanower Hossain, Md Mahfuzul Islam, Md Hasan Ali, Munira Mohtasim, Naureen Sharmi, and Mahamuda Afroze for their support and reviewing this manuscript.
| 1. | Hadi F, Awan SJ, Tayyeb A, Maqbool T, Shehzadi S, Malik S, Kausar H, Malik A. Hepato-protective role of itraconazole mediated cytochrome p450 pathway inhibition in liver fibrosis. Pak J Pharm Sci. 2020;33:2751-2758. [PubMed] [DOI] [Full Text] |
| 2. | Peng W, Cheng S, Bao Z, Wang Y, Zhou W, Wang J, Yang Q, Chen C, Wang W. Advances in the research of nanodrug delivery system for targeted treatment of liver fibrosis. Biomed Pharmacother. 2021;137:111342. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 7] [Cited by in RCA: 28] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
| 3. | Panagiotakopoulos L, Sandul AL; DHSc, Conners EE, Foster MA, Nelson NP, Wester C; Collaborators. CDC Recommendations for Hepatitis C Testing Among Perinatally Exposed Infants and Children - United States, 2023. MMWR Recomm Rep. 2023;72:1-21. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 30] [Reference Citation Analysis (0)] |
| 4. | Macdonald S, Andreola F, Bachtiger P, Amoros A, Pavesi M, Mookerjee R, Zheng YB, Gronbaek H, Gerbes AL, Sola E, Caraceni P, Moreau R, Gines P, Arroyo V, Jalan R. Cell death markers in patients with cirrhosis and acute decompensation. Hepatology. 2018;67:989-1002. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 55] [Cited by in RCA: 75] [Article Influence: 10.7] [Reference Citation Analysis (0)] |
| 5. | Roehlen N, Crouchet E, Baumert TF. Liver Fibrosis: Mechanistic Concepts and Therapeutic Perspectives. Cells. 2020;9:875. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 198] [Cited by in RCA: 801] [Article Influence: 160.2] [Reference Citation Analysis (0)] |
| 6. | Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther. 2025;10:33. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 5] [Cited by in RCA: 34] [Article Influence: 34.0] [Reference Citation Analysis (0)] |
| 7. | Moon AM, Singal AG, Tapper EB. Contemporary Epidemiology of Chronic Liver Disease and Cirrhosis. Clin Gastroenterol Hepatol. 2020;18:2650-2666. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 805] [Cited by in RCA: 754] [Article Influence: 150.8] [Reference Citation Analysis (0)] |
| 8. | Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol. 2019;70:151-171. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1382] [Cited by in RCA: 2368] [Article Influence: 394.7] [Reference Citation Analysis (0)] |
| 9. | Rahman SR, Roper JA, Grove JI, Aithal GP, Pun KT, Bennett AJ. Integrins as a drug target in liver fibrosis. Liver Int. 2022;42:507-521. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 46] [Article Influence: 15.3] [Reference Citation Analysis (0)] |
| 10. | Zhang J, Liu Q, He J, Li Y. Novel Therapeutic Targets in Liver Fibrosis. Front Mol Biosci. 2021;8:766855. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 34] [Article Influence: 8.5] [Reference Citation Analysis (0)] |
| 11. | Liu Z, Zhu P, Zhang L, Xiong B, Tao J, Guan W, Li C, Chen C, Gu J, Duanmu J, Zhang W. Autophagy inhibition attenuates the induction of anti-inflammatory effect of catalpol in liver fibrosis. Biomed Pharmacother. 2018;103:1262-1271. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 35] [Cited by in RCA: 63] [Article Influence: 9.0] [Reference Citation Analysis (0)] |
| 12. | Zhang S, Wang Z, Zhu J, Xu T, Zhao Y, Zhao H, Tang F, Li Z, Zhou J, Gao D, Tian X, Yao J. Carnosic Acid Alleviates BDL-Induced Liver Fibrosis through miR-29b-3p-Mediated Inhibition of the High-Mobility Group Box 1/Toll-Like Receptor 4 Signaling Pathway in Rats. Front Pharmacol. 2017;8:976. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 15] [Cited by in RCA: 26] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
| 13. | Raj D, Sharma V, Upadhyaya A, Kumar N, Joshi R, Acharya V, Kumar D, Patial V. Swertia purpurascens Wall ethanolic extract mitigates hepatic fibrosis and restores hepatic hepcidin levels via inhibition of TGFβ/SMAD/NFκB signaling in rats. J Ethnopharmacol. 2022;284:114741. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 6] [Cited by in RCA: 8] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
| 14. | Zhang H, Chen Q, Dahan A, Xue J, Wei L, Tan W, Zhang G. Transcriptomic analyses reveal the molecular mechanisms of schisandrin B alleviates CCl(4)-induced liver fibrosis in rats by RNA-sequencing. Chem Biol Interact. 2019;309:108675. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 28] [Article Influence: 4.7] [Reference Citation Analysis (0)] |
| 15. | Milito A, Brancaccio M, D'Argenio G, Castellano I. Natural Sulfur-Containing Compounds: An Alternative Therapeutic Strategy against Liver Fibrosis. Cells. 2019;8:0. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 31] [Cited by in RCA: 52] [Article Influence: 8.7] [Reference Citation Analysis (0)] |
| 16. | Alcolado R, Arthur MJ, Iredale JP. Pathogenesis of liver fibrosis. Clin Sci (Lond). 1997;92:103-112. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 123] [Cited by in RCA: 131] [Article Influence: 4.7] [Reference Citation Analysis (0)] |
| 17. | Casini A, Ceni E, Salzano R, Biondi P, Parola M, Galli A, Foschi M, Caligiuri A, Pinzani M, Surrenti C. Neutrophil-derived superoxide anion induces lipid peroxidation and stimulates collagen synthesis in human hepatic stellate cells: role of nitric oxide. Hepatology. 1997;25:361-367. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 16] [Cited by in RCA: 68] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
| 18. | Ghatak S, Biswas A, Dhali GK, Chowdhury A, Boyer JL, Santra A. Oxidative stress and hepatic stellate cell activation are key events in arsenic induced liver fibrosis in mice. Toxicol Appl Pharmacol. 2011;251:59-69. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 111] [Cited by in RCA: 121] [Article Influence: 8.6] [Reference Citation Analysis (0)] |
| 19. | Mederacke I, Hsu CC, Troeger JS, Huebener P, Mu X, Dapito DH, Pradere JP, Schwabe RF. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun. 2013;4:2823. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 750] [Cited by in RCA: 1078] [Article Influence: 98.0] [Reference Citation Analysis (0)] |
| 20. | Novo E, di Bonzo LV, Cannito S, Colombatto S, Parola M. Hepatic myofibroblasts: a heterogeneous population of multifunctional cells in liver fibrogenesis. Int J Biochem Cell Biol. 2009;41:2089-2093. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 78] [Cited by in RCA: 80] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 21. | Luedde T, Kaplowitz N, Schwabe RF. Cell death and cell death responses in liver disease: mechanisms and clinical relevance. Gastroenterology. 2014;147:765-783.e4. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 456] [Cited by in RCA: 588] [Article Influence: 53.5] [Reference Citation Analysis (0)] |
| 22. | Seki E, Schwabe RF. Hepatic inflammation and fibrosis: functional links and key pathways. Hepatology. 2015;61:1066-1079. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 548] [Cited by in RCA: 755] [Article Influence: 75.5] [Reference Citation Analysis (0)] |
| 23. | Itaba N, Kono Y, Watanabe K, Yokobata T, Oka H, Osaki M, Kakuta H, Morimoto M, Shiota G. Reversal of established liver fibrosis by IC-2-engineered mesenchymal stem cell sheets. Sci Rep. 2019;9:6841. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 17] [Cited by in RCA: 19] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
| 24. | Sapisochin G, Bruix J. Liver transplantation for hepatocellular carcinoma: outcomes and novel surgical approaches. Nat Rev Gastroenterol Hepatol. 2017;14:203-217. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 221] [Cited by in RCA: 346] [Article Influence: 43.3] [Reference Citation Analysis (0)] |
| 25. | Horwitz JK, Agopian VG. Indication of Liver Transplant for HCC: Current Status and Future Directions. Curr Hepatology Rep. 2024;23:185-192. [RCA] [DOI] [Full Text] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 26. | Lucey MR, Furuya KN, Foley DP. Liver Transplantation. N Engl J Med. 2023;389:1888-1900. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 68] [Cited by in RCA: 76] [Article Influence: 38.0] [Reference Citation Analysis (0)] |
| 27. | Waheed A, Gul MH, Shabbir MUB, Raja HAA, Wardak AB, Hussaini H, Shah Y. FDA approves Resmetirom: groundbreaking treatment for NASH liver scarring in moderate to advanced fibrosis. Ann Med Surg (Lond). 2025;87:1088-1091. [PubMed] [DOI] [Full Text] |
| 28. | Shakeel L, Shaukat A, Akilimali A. Resmetirom: A Breakthrough in the Treatment of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). Health Sci Rep. 2025;8:e70920. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 3] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 29. | Mousa AM, Mahmoud M, AlShuraiaan GM. Resmetirom: The First Disease-Specific Treatment for MASH. Int J Endocrinol. 2025;2025:6430023. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 9] [Cited by in RCA: 6] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
| 30. | Ali M, Khan T, Fatima K, Ali QUA, Ovais M, Khalil AT, Ullah I, Raza A, Shinwari ZK, Idrees M. Selected hepatoprotective herbal medicines: Evidence from ethnomedicinal applications, animal models, and possible mechanism of actions. Phytother Res. 2018;32:199-215. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 41] [Cited by in RCA: 77] [Article Influence: 9.6] [Reference Citation Analysis (0)] |
| 31. | Das R, Mitra S, Tareq AM, Emran TB, Hossain MJ, Alqahtani AM, Alghazwani Y, Dhama K, Simal-Gandara J. Medicinal plants used against hepatic disorders in Bangladesh: A comprehensive review. J Ethnopharmacol. 2022;282:114588. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 55] [Cited by in RCA: 40] [Article Influence: 13.3] [Reference Citation Analysis (0)] |
| 32. | Li J, Hu R, Xu S, Li Y, Qin Y, Wu Q, Xiao Z. Xiaochaihutang attenuates liver fibrosis by activation of Nrf2 pathway in rats. Biomed Pharmacother. 2017;96:847-853. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 23] [Cited by in RCA: 41] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
| 33. | Zhao XA, Chen G, Liu Y, Chen Y, Wu H, Xiong Y, Wang G, Jia B, Li Y, Xia J, Wang J, Yan X, Zhang Z, Huang R, Wu C. Curcumin reduces Ly6C(hi) monocyte infiltration to protect against liver fibrosis by inhibiting Kupffer cells activation to reduce chemokines secretion. Biomed Pharmacother. 2018;106:868-878. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 37] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
| 34. | Zhang F, Zhang Z, Chen L, Kong D, Zhang X, Lu C, Lu Y, Zheng S. Curcumin attenuates angiogenesis in liver fibrosis and inhibits angiogenic properties of hepatic stellate cells. J Cell Mol Med. 2014;18:1392-1406. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 100] [Cited by in RCA: 114] [Article Influence: 10.4] [Reference Citation Analysis (0)] |
| 35. | Wang FD, Zhou J, Chen EQ. Molecular Mechanisms and Potential New Therapeutic Drugs for Liver Fibrosis. Front Pharmacol. 2022;13:787748. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 33] [Cited by in RCA: 34] [Article Influence: 11.3] [Reference Citation Analysis (0)] |
| 36. | Bijak M. Silybin, a Major Bioactive Component of Milk Thistle (Silybum marianum L. Gaernt.)-Chemistry, Bioavailability, and Metabolism. Molecules. 2017;22:1942. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 212] [Cited by in RCA: 238] [Article Influence: 29.8] [Reference Citation Analysis (0)] |
| 37. | Abenavoli L, Izzo AA, Milić N, Cicala C, Santini A, Capasso R. Milk thistle (Silybum marianum): A concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother Res. 2018;32:2202-2213. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 245] [Cited by in RCA: 270] [Article Influence: 38.6] [Reference Citation Analysis (0)] |
| 38. | Mohanty C, Sahoo SK. The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials. 2010;31:6597-6611. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 340] [Cited by in RCA: 379] [Article Influence: 25.3] [Reference Citation Analysis (0)] |
| 39. | Liu Y, Zheng Y, Yang Y, Liu K, Wu J, Gao P, Zhang C. Exosomes in liver fibrosis: The role of modulating hepatic stellate cells and immune cells, and prospects for clinical applications. Front Immunol. 2023;14:1133297. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 24] [Reference Citation Analysis (0)] |
| 40. | Chiabotto G, Ceccotti E, Tapparo M, Camussi G, Bruno S. Human Liver Stem Cell-Derived Extracellular Vesicles Target Hepatic Stellate Cells and Attenuate Their Pro-fibrotic Phenotype. Front Cell Dev Biol. 2021;9:777462. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 28] [Article Influence: 7.0] [Reference Citation Analysis (0)] |
| 41. | Chiabotto G, Semnani A, Ceccotti E, Guenza M, Camussi G, Bruno S. Mesenchymal Stromal Cell-Derived Extracellular Vesicles for Reversing Hepatic Fibrosis in 3D Liver Spheroids. Biomedicines. 2024;12:1849. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 3] [Reference Citation Analysis (0)] |
| 42. | Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK. Extracellular matrix structure. Adv Drug Deliv Rev. 2016;97:4-27. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1153] [Cited by in RCA: 1652] [Article Influence: 183.6] [Reference Citation Analysis (0)] |
| 43. | Villesen IF, Daniels SJ, Leeming DJ, Karsdal MA, Nielsen MJ. Review article: the signalling and functional role of the extracellular matrix in the development of liver fibrosis. Aliment Pharmacol Ther. 2020;52:85-97. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 45] [Article Influence: 9.0] [Reference Citation Analysis (1)] |
| 44. | Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol. 2011;6:425-456. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1096] [Cited by in RCA: 1392] [Article Influence: 99.4] [Reference Citation Analysis (0)] |
| 45. | Zhang Y, Yu X, Wang Z, Yin M, Zhao Z, Li Y, Li W. Pokeweed antiviral protein attenuates liver fibrosis in mice through regulating Wnt/Jnk mediated glucose metabolism. Saudi J Gastroenterol. 2018;24:157-164. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 12] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
| 46. | Wu J, Xue X, Fan G, Gu Y, Zhou F, Zheng Q, Liu R, Li Y, Ma B, Li S, Huang G, Ma L, Li X. Ferulic Acid Ameliorates Hepatic Inflammation and Fibrotic Liver Injury by Inhibiting PTP1B Activity and Subsequent Promoting AMPK Phosphorylation. Front Pharmacol. 2021;12:754976. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 6] [Cited by in RCA: 31] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
| 47. | Karimi J, Mohammadalipour A, Sheikh N, Khodadadi I, Hashemnia M, Goudarzi F, Khanjarsim V, Solgi G, Hajilooi M, Bahabadi M, Kheiripour N, Hedayatyanfard K. Protective effects of combined Losartan and Nilotinib on carbon tetrachloride (CCl(4))-induced liver fibrosis in rats. Drug Chem Toxicol. 2020;43:468-478. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 18] [Article Influence: 2.6] [Reference Citation Analysis (0)] |
| 48. | Wehr A, Baeck C, Heymann F, Niemietz PM, Hammerich L, Martin C, Zimmermann HW, Pack O, Gassler N, Hittatiya K, Ludwig A, Luedde T, Trautwein C, Tacke F. Chemokine receptor CXCR6-dependent hepatic NK T Cell accumulation promotes inflammation and liver fibrosis. J Immunol. 2013;190:5226-5236. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 217] [Cited by in RCA: 219] [Article Influence: 18.3] [Reference Citation Analysis (0)] |
| 49. | Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, Jang MK, Guenther ND, Mederacke I, Friedman R, Dragomir AC, Aloman C, Schwabe RF. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology. 2013;58:1461-1473. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 375] [Cited by in RCA: 475] [Article Influence: 39.6] [Reference Citation Analysis (0)] |
| 50. | Marrone G, Shah VH, Gracia-Sancho J. Sinusoidal communication in liver fibrosis and regeneration. J Hepatol. 2016;65:608-617. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 187] [Cited by in RCA: 250] [Article Influence: 27.8] [Reference Citation Analysis (0)] |
| 51. | Canbay A, Higuchi H, Bronk SF, Taniai M, Sebo TJ, Gores GJ. Fas enhances fibrogenesis in the bile duct ligated mouse: a link between apoptosis and fibrosis. Gastroenterology. 2002;123:1323-1330. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 234] [Cited by in RCA: 227] [Article Influence: 9.9] [Reference Citation Analysis (0)] |
| 52. | Elpek GÖ. Cellular and molecular mechanisms in the pathogenesis of liver fibrosis: An update. World J Gastroenterol. 2014;20:7260-7276. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 289] [Cited by in RCA: 297] [Article Influence: 27.0] [Reference Citation Analysis (4)] |
| 53. | Schulze RJ, Schott MB, Casey CA, Tuma PL, McNiven MA. The cell biology of the hepatocyte: A membrane trafficking machine. J Cell Biol. 2019;218:2096-2112. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 125] [Cited by in RCA: 133] [Article Influence: 22.2] [Reference Citation Analysis (0)] |
| 54. | An P, Wei LL, Zhao S, Sverdlov DY, Vaid KA, Miyamoto M, Kuramitsu K, Lai M, Popov YV. Hepatocyte mitochondria-derived danger signals directly activate hepatic stellate cells and drive progression of liver fibrosis. Nat Commun. 2020;11:2362. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 71] [Cited by in RCA: 218] [Article Influence: 43.6] [Reference Citation Analysis (1)] |
| 55. | Gaul S, Leszczynska A, Alegre F, Kaufmann B, Johnson CD, Adams LA, Wree A, Damm G, Seehofer D, Calvente CJ, Povero D, Kisseleva T, Eguchi A, McGeough MD, Hoffman HM, Pelegrin P, Laufs U, Feldstein AE. Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis. J Hepatol. 2021;74:156-167. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 260] [Cited by in RCA: 405] [Article Influence: 101.3] [Reference Citation Analysis (0)] |
| 56. | Li J, Zeng C, Zheng B, Liu C, Tang M, Jiang Y, Chang Y, Song W, Wang Y, Yang C. HMGB1-induced autophagy facilitates hepatic stellate cells activation: a new pathway in liver fibrosis. Clin Sci (Lond). 2018;132:1645-1667. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 35] [Cited by in RCA: 60] [Article Influence: 8.6] [Reference Citation Analysis (0)] |
| 57. | Luo X, Xu ZX, Wu JC, Luo SZ, Xu MY. Hepatocyte-derived exosomal miR-27a activateshepatic stellate cells through the inhibitionof PINK1-mediated mitophagy in MAFLD. Mol Ther Nucleic Acids. 2021;26:1241-1254. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 43] [Article Influence: 10.8] [Reference Citation Analysis (1)] |
| 58. | Wynn TA, Barron L. Macrophages: master regulators of inflammation and fibrosis. Semin Liver Dis. 2010;30:245-257. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1127] [Cited by in RCA: 1088] [Article Influence: 72.5] [Reference Citation Analysis (0)] |
| 59. | Karlmark KR, Weiskirchen R, Zimmermann HW, Gassler N, Ginhoux F, Weber C, Merad M, Luedde T, Trautwein C, Tacke F. Hepatic recruitment of the inflammatory Gr1+ monocyte subset upon liver injury promotes hepatic fibrosis. Hepatology. 2009;50:261-274. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 640] [Cited by in RCA: 634] [Article Influence: 39.6] [Reference Citation Analysis (0)] |
| 60. | Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18:151-166. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 274] [Cited by in RCA: 1267] [Article Influence: 316.8] [Reference Citation Analysis (0)] |
| 61. | Weiskirchen R, Weiskirchen S, Tacke F. Recent advances in understanding liver fibrosis: bridging basic science and individualized treatment concepts. F1000Res. 2018;7:F1000 Faculty Rev-F1000 Faculty 921. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 71] [Cited by in RCA: 78] [Article Influence: 11.1] [Reference Citation Analysis (0)] |
| 62. | Jaeschke H. Reactive oxygen and mechanisms of inflammatory liver injury: Present concepts. J Gastroenterol Hepatol. 2011;26 Suppl 1:173-179. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 361] [Cited by in RCA: 418] [Article Influence: 29.9] [Reference Citation Analysis (0)] |
| 63. | Abdelghffar EA, Obaid WA, Alamoudi MO, Mohammedsaleh ZM, Annaz H, Abdelfattah MAO, Sobeh M. Thymus fontanesii attenuates CCl(4)-induced oxidative stress and inflammation in mild liver fibrosis. Biomed Pharmacother. 2022;148:112738. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 11] [Article Influence: 3.7] [Reference Citation Analysis (0)] |
| 64. | Liu C, Tao Q, Sun M, Wu JZ, Yang W, Jian P, Peng J, Hu Y, Liu C, Liu P. Kupffer cells are associated with apoptosis, inflammation and fibrotic effects in hepatic fibrosis in rats. Lab Invest. 2010;90:1805-1816. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 94] [Cited by in RCA: 115] [Article Influence: 7.7] [Reference Citation Analysis (0)] |
| 65. | Ding BS, Nolan DJ, Butler JM, James D, Babazadeh AO, Rosenwaks Z, Mittal V, Kobayashi H, Shido K, Lyden D, Sato TN, Rabbany SY, Rafii S. Inductive angiocrine signals from sinusoidal endothelium are required for liver regeneration. Nature. 2010;468:310-315. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 700] [Cited by in RCA: 650] [Article Influence: 43.3] [Reference Citation Analysis (0)] |
| 66. | Xie G, Wang X, Wang L, Wang L, Atkinson RD, Kanel GC, Gaarde WA, Deleve LD. Role of differentiation of liver sinusoidal endothelial cells in progression and regression of hepatic fibrosis in rats. Gastroenterology. 2012;142:918-927.e6. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 279] [Cited by in RCA: 306] [Article Influence: 23.5] [Reference Citation Analysis (0)] |
| 67. | Deleve LD, Wang X, Guo Y. Sinusoidal endothelial cells prevent rat stellate cell activation and promote reversion to quiescence. Hepatology. 2008;48:920-930. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 321] [Cited by in RCA: 301] [Article Influence: 17.7] [Reference Citation Analysis (0)] |
| 68. | Poisson J, Lemoinne S, Boulanger C, Durand F, Moreau R, Valla D, Rautou PE. Liver sinusoidal endothelial cells: Physiology and role in liver diseases. J Hepatol. 2017;66:212-227. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 475] [Cited by in RCA: 728] [Article Influence: 91.0] [Reference Citation Analysis (0)] |
| 69. | Wei M, Zhang Y, Zhang H, Huang Z, Miao H, Zhang T, Lu B, Ji L. HMGB1 induced endothelial to mesenchymal transition in liver fibrosis: The key regulation of early growth response factor 1. Biochim Biophys Acta Gen Subj. 2022;1866:130202. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 17] [Cited by in RCA: 14] [Article Influence: 4.7] [Reference Citation Analysis (0)] |
| 70. | Kaftanovskaya EM, Ng HH, Soula M, Rivas B, Myhr C, Ho BA, Cervantes BA, Shupe TD, Devarasetty M, Hu X, Xu X, Patnaik S, Wilson KJ, Barnaeva E, Ferrer M, Southall NT, Marugan JJ, Bishop CE, Agoulnik IU, Agoulnik AI. Therapeutic effects of a small molecule agonist of the relaxin receptor ML290 in liver fibrosis. FASEB J. 2019;33:12435-12446. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 9] [Cited by in RCA: 26] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
| 71. | Carloni V, Romanelli RG, Pinzani M, Laffi G, Gentilini P. Expression and function of integrin receptors for collagen and laminin in cultured human hepatic stellate cells. Gastroenterology. 1996;110:1127-1136. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 86] [Cited by in RCA: 82] [Article Influence: 2.8] [Reference Citation Analysis (0)] |
| 72. | Roeb E, Purucker E, Breuer B, Nguyen H, Heinrich PC, Rose-John S, Matern S. TIMP expression in toxic and cholestatic liver injury in rat. J Hepatol. 1997;27:535-544. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 63] [Cited by in RCA: 65] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
| 73. | Benyon RC, Arthur MJ. Extracellular matrix degradation and the role of hepatic stellate cells. Semin Liver Dis. 2001;21:373-384. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 358] [Cited by in RCA: 371] [Article Influence: 15.5] [Reference Citation Analysis (0)] |
| 74. | Geerts A. History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells. Semin Liver Dis. 2001;21:311-335. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 548] [Cited by in RCA: 567] [Article Influence: 23.6] [Reference Citation Analysis (0)] |
| 75. | Yoneda A, Sakai-Sawada K, Niitsu Y, Tamura Y. Vitamin A and insulin are required for the maintenance of hepatic stellate cell quiescence. Exp Cell Res. 2016;341:8-17. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 25] [Cited by in RCA: 39] [Article Influence: 4.3] [Reference Citation Analysis (0)] |
| 76. | Murphy FR, Issa R, Zhou X, Ratnarajah S, Nagase H, Arthur MJ, Benyon C, Iredale JP. Inhibition of apoptosis of activated hepatic stellate cells by tissue inhibitor of metalloproteinase-1 is mediated via effects on matrix metalloproteinase inhibition: implications for reversibility of liver fibrosis. J Biol Chem. 2002;277:11069-11076. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 328] [Cited by in RCA: 345] [Article Influence: 15.0] [Reference Citation Analysis (0)] |
| 77. | Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrotic diseases: nearing the starting line. Sci Transl Med. 2013;5:167sr1. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 455] [Cited by in RCA: 480] [Article Influence: 40.0] [Reference Citation Analysis (0)] |
| 78. | Iwaisako K, Jiang C, Zhang M, Cong M, Moore-Morris TJ, Park TJ, Liu X, Xu J, Wang P, Paik YH, Meng F, Asagiri M, Murray LA, Hofmann AF, Iida T, Glass CK, Brenner DA, Kisseleva T. Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci U S A. 2014;111:E3297-E3305. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 309] [Cited by in RCA: 417] [Article Influence: 37.9] [Reference Citation Analysis (0)] |
| 79. | Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017;14:397-411. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1221] [Cited by in RCA: 2087] [Article Influence: 260.9] [Reference Citation Analysis (0)] |
| 80. | Derynck R, Zhang YE. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature. 2003;425:577-584. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3797] [Cited by in RCA: 4355] [Article Influence: 198.0] [Reference Citation Analysis (1)] |
| 81. | Inagaki Y, Okazaki I. Emerging insights into Transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut. 2007;56:284-292. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 339] [Cited by in RCA: 408] [Article Influence: 22.7] [Reference Citation Analysis (0)] |
| 82. | Arroyo N, Villamayor L, Díaz I, Carmona R, Ramos-Rodríguez M, Muñoz-Chápuli R, Pasquali L, Toscano MG, Martín F, Cano DA, Rojas A. GATA4 induces liver fibrosis regression by deactivating hepatic stellate cells. JCI Insight. 2021;6:e150059. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 29] [Article Influence: 7.3] [Reference Citation Analysis (0)] |
| 83. | Friedman SL. Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol Rev. 2008;88:125-172. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2244] [Cited by in RCA: 2218] [Article Influence: 130.5] [Reference Citation Analysis (0)] |
| 84. | Liu QQ, Chen J, Ma T, Huang W, Lu CH. DCDC2 inhibits hepatic stellate cell activation and ameliorates CCl(4)-induced liver fibrosis by suppressing Wnt/β-catenin signaling. Sci Rep. 2024;14:9425. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 7] [Reference Citation Analysis (0)] |
| 85. | Ge WS, Wang YJ, Wu JX, Fan JG, Chen YW, Zhu L. β-catenin is overexpressed in hepatic fibrosis and blockage of Wnt/β-catenin signaling inhibits hepatic stellate cell activation. Mol Med Rep. 2014;9:2145-2151. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 89] [Cited by in RCA: 96] [Article Influence: 8.7] [Reference Citation Analysis (0)] |
| 86. | Luedde T, Schwabe RF. NF-κB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol. 2011;8:108-118. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1094] [Cited by in RCA: 1109] [Article Influence: 79.2] [Reference Citation Analysis (0)] |
| 87. | Oakley F, Mann J, Nailard S, Smart DE, Mungalsingh N, Constandinou C, Ali S, Wilson SJ, Millward-Sadler H, Iredale JP, Mann DA. Nuclear factor-kappaB1 (p50) limits the inflammatory and fibrogenic responses to chronic injury. Am J Pathol. 2005;166:695-708. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 93] [Cited by in RCA: 102] [Article Influence: 5.1] [Reference Citation Analysis (0)] |
| 88. | Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18:6853-6866. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3079] [Cited by in RCA: 3123] [Article Influence: 120.1] [Reference Citation Analysis (3)] |
| 89. | Hernández-Muñoz I, de la Torre P, Sánchez-Alcázar JA, García I, Santiago E, Muñoz-Yagüe MT, Solís-Herruzo JA. Tumor necrosis factor alpha inhibits collagen alpha 1(I) gene expression in rat hepatic stellate cells through a G protein. Gastroenterology. 1997;113:625-640. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 47] [Cited by in RCA: 44] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
| 90. | Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001;410:37-40. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3924] [Cited by in RCA: 4019] [Article Influence: 167.5] [Reference Citation Analysis (33)] |
| 91. | Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001;81:807-869. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2510] [Cited by in RCA: 2564] [Article Influence: 106.8] [Reference Citation Analysis (0)] |
| 92. | Davis RJ. Signal transduction by the JNK group of MAP kinases. Cell. 2000;103:239-252. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3252] [Cited by in RCA: 3357] [Article Influence: 134.3] [Reference Citation Analysis (0)] |
| 93. | Santos SD, Verveer PJ, Bastiaens PI. Growth factor-induced MAPK network topology shapes Erk response determining PC-12 cell fate. Nat Cell Biol. 2007;9:324-330. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 509] [Cited by in RCA: 481] [Article Influence: 26.7] [Reference Citation Analysis (0)] |
| 94. | Luca VC, Kim BC, Ge C, Kakuda S, Wu D, Roein-Peikar M, Haltiwanger RS, Zhu C, Ha T, Garcia KC. Notch-Jagged complex structure implicates a catch bond in tuning ligand sensitivity. Science. 2017;355:1320-1324. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 179] [Cited by in RCA: 235] [Article Influence: 29.4] [Reference Citation Analysis (0)] |
| 95. | Yimlamai D, Christodoulou C, Galli GG, Yanger K, Pepe-Mooney B, Gurung B, Shrestha K, Cahan P, Stanger BZ, Camargo FD. Hippo pathway activity influences liver cell fate. Cell. 2014;157:1324-1338. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 556] [Cited by in RCA: 678] [Article Influence: 61.6] [Reference Citation Analysis (0)] |
| 96. | Yanger K, Zong Y, Maggs LR, Shapira SN, Maddipati R, Aiello NM, Thung SN, Wells RG, Greenbaum LE, Stanger BZ. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes Dev. 2013;27:719-724. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 340] [Cited by in RCA: 391] [Article Influence: 32.6] [Reference Citation Analysis (0)] |
| 97. | Groth C, Alvord WG, Quiñones OA, Fortini ME. Pharmacological analysis of Drosophila melanogaster gamma-secretase with respect to differential proteolysis of Notch and APP. Mol Pharmacol. 2010;77:567-574. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 20] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 98. | Hong W, Zhang G, Lu H, Guo Y, Zheng S, Zhu H, Xiao Y, Papa APD, Wu C, Sun L, Chen B, Bai Y. Epithelial and interstitial Notch1 activity contributes to the myofibroblastic phenotype and fibrosis. Cell Commun Signal. 2019;17:145. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 10] [Cited by in RCA: 28] [Article Influence: 4.7] [Reference Citation Analysis (0)] |
| 99. | Pratap A, Singh S, Mundra V, Yang N, Panakanti R, Eason JD, Mahato RI. Attenuation of early liver fibrosis by pharmacological inhibition of smoothened receptor signaling. J Drug Target. 2012;20:770-782. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 27] [Cited by in RCA: 39] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 100. | Zhang F, Hao M, Jin H, Yao Z, Lian N, Wu L, Shao J, Chen A, Zheng S. Canonical hedgehog signalling regulates hepatic stellate cell-mediated angiogenesis in liver fibrosis. Br J Pharmacol. 2017;174:409-423. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 47] [Cited by in RCA: 65] [Article Influence: 8.1] [Reference Citation Analysis (0)] |
| 101. | Gao L, Zhang Z, Zhang P, Yu M, Yang T. Role of canonical Hedgehog signaling pathway in liver. Int J Biol Sci. 2018;14:1636-1644. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 36] [Cited by in RCA: 61] [Article Influence: 8.7] [Reference Citation Analysis (0)] |
| 102. | Yu FX, Zhao B, Guan KL. Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell. 2015;163:811-828. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1241] [Cited by in RCA: 1746] [Article Influence: 194.0] [Reference Citation Analysis (0)] |
| 103. | Gong J, Mou T, Wu H, Wu Z. Brg1 regulates murine liver regeneration by targeting miR-187-5p dependent on Hippo signalling pathway. J Cell Mol Med. 2020;24:11592-11602. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 11] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
| 104. | Ding N, Hah N, Yu RT, Sherman MH, Benner C, Leblanc M, He M, Liddle C, Downes M, Evans RM. BRD4 is a novel therapeutic target for liver fibrosis. Proc Natl Acad Sci U S A. 2015;112:15713-15718. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 128] [Cited by in RCA: 170] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
| 105. | Zhubanchaliyev A, Temirbekuly A, Kongrtay K, Wanshura LC, Kunz J. Targeting Mechanotransduction at the Transcriptional Level: YAP and BRD4 Are Novel Therapeutic Targets for the Reversal of Liver Fibrosis. Front Pharmacol. 2016;7:462. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 26] [Cited by in RCA: 44] [Article Influence: 4.9] [Reference Citation Analysis (0)] |
| 106. | Liu-Chittenden Y, Huang B, Shim JS, Chen Q, Lee SJ, Anders RA, Liu JO, Pan D. Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP. Genes Dev. 2012;26:1300-1305. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 892] [Cited by in RCA: 1202] [Article Influence: 92.5] [Reference Citation Analysis (0)] |
| 107. | Pinzani M, Gesualdo L, Sabbah GM, Abboud HE. Effects of platelet-derived growth factor and other polypeptide mitogens on DNA synthesis and growth of cultured rat liver fat-storing cells. J Clin Invest. 1989;84:1786-1793. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 370] [Cited by in RCA: 380] [Article Influence: 10.6] [Reference Citation Analysis (0)] |
| 108. | Schnabl B, Bradham CA, Bennett BL, Manning AM, Stefanovic B, Brenner DA. TAK1/JNK and p38 have opposite effects on rat hepatic stellate cells. Hepatology. 2001;34:953-963. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 100] [Cited by in RCA: 101] [Article Influence: 4.2] [Reference Citation Analysis (0)] |
| 109. | Luo K. Signaling Cross Talk between TGF-β/Smad and Other Signaling Pathways. Cold Spring Harb Perspect Biol. 2017;9:a022137. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 415] [Cited by in RCA: 459] [Article Influence: 57.4] [Reference Citation Analysis (0)] |
| 110. | Chan YT, Wang N, Tan HY, Li S, Feng Y. Targeting Hepatic Stellate Cells for the Treatment of Liver Fibrosis by Natural Products: Is It the Dawning of a New Era? Front Pharmacol. 2020;11:548. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 39] [Cited by in RCA: 39] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
| 111. | Russell JO, Monga SP. Wnt/β-Catenin Signaling in Liver Development, Homeostasis, and Pathobiology. Annu Rev Pathol. 2018;13:351-378. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 178] [Cited by in RCA: 362] [Article Influence: 45.3] [Reference Citation Analysis (0)] |
| 112. | Yu J, Hu Y, Gao Y, Li Q, Zeng Z, Li Y, Chen H. Kindlin-2 regulates hepatic stellate cells activation and liver fibrogenesis. Cell Death Discov. 2018;4:34. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 11] [Cited by in RCA: 21] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 113. | Zhang Y, Ren L, Tian Y, Guo X, Wei F, Zhang Y. Signaling pathways that activate hepatic stellate cells during liver fibrosis. Front Med (Lausanne). 2024;11:1454980. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 6] [Reference Citation Analysis (0)] |
| 114. | Fabregat I, Caballero-Díaz D. Transforming Growth Factor-β-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front Oncol. 2018;8:357. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 150] [Cited by in RCA: 263] [Article Influence: 37.6] [Reference Citation Analysis (0)] |
| 115. | Mayorca-Guiliani AE, Leeming DJ, Henriksen K, Mortensen JH, Nielsen SH, Anstee QM, Sanyal AJ, Karsdal MA, Schuppan D. ECM formation and degradation during fibrosis, repair, and regeneration. NPJ Metab Health Dis. 2025;3:25. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 10] [Article Influence: 10.0] [Reference Citation Analysis (0)] |
| 116. | Massagué J, Sheppard D. TGF-β signaling in health and disease. Cell. 2023;186:4007-4037. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 12] [Cited by in RCA: 426] [Article Influence: 213.0] [Reference Citation Analysis (0)] |
| 117. | Somanader DVN, Zhao P, Widdop RE, Samuel CS. The involvement of the Wnt/β-catenin signaling cascade in fibrosis progression and its therapeutic targeting by relaxin. Biochem Pharmacol. 2024;223:116130. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 17] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
| 118. | Zhou B, Liu Y, Kahn M, Ann DK, Han A, Wang H, Nguyen C, Flodby P, Zhong Q, Krishnaveni MS, Liebler JM, Minoo P, Crandall ED, Borok Z. Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP). J Biol Chem. 2012;287:7026-7038. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 189] [Cited by in RCA: 221] [Article Influence: 17.0] [Reference Citation Analysis (0)] |
| 119. | Akhmetshina A, Palumbo K, Dees C, Bergmann C, Venalis P, Zerr P, Horn A, Kireva T, Beyer C, Zwerina J, Schneider H, Sadowski A, Riener MO, MacDougald OA, Distler O, Schett G, Distler JH. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat Commun. 2012;3:735. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 532] [Cited by in RCA: 692] [Article Influence: 53.2] [Reference Citation Analysis (0)] |
| 120. | Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther. 2024;9:53. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 655] [Cited by in RCA: 753] [Article Influence: 753.0] [Reference Citation Analysis (0)] |
| 121. | Zhang C, Sun C, Zhao Y, Ye B, Yu G. Signaling pathways of liver regeneration: Biological mechanisms and implications. iScience. 2024;27:108683. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 16] [Reference Citation Analysis (0)] |
| 122. | Mitropoulou G, Kompoura V, Saffioti F, Mavroeidis VK. The Role of Matrix Metalloproteinases in Liver Function and Disease. Front Biosci (Landmark Ed). 2025;30:27127. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 123. | Manicone AM, McGuire JK. Matrix metalloproteinases as modulators of inflammation. Semin Cell Dev Biol. 2008;19:34-41. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 446] [Cited by in RCA: 428] [Article Influence: 25.2] [Reference Citation Analysis (0)] |
| 124. | Gungor MZ, Uysal M, Senturk S. The Bright and the Dark Side of TGF-β Signaling in Hepatocellular Carcinoma: Mechanisms, Dysregulation, and Therapeutic Implications. Cancers (Basel). 2022;14:940. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 35] [Article Influence: 11.7] [Reference Citation Analysis (0)] |
| 125. | Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009;19:128-139. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1451] [Cited by in RCA: 1446] [Article Influence: 90.4] [Reference Citation Analysis (4)] |
| 126. | Novosyadlyy R, Dudas J, Pannem R, Ramadori G, Scharf JG. Crosstalk between PDGF and IGF-I receptors in rat liver myofibroblasts: implication for liver fibrogenesis. Lab Invest. 2006;86:710-723. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 21] [Cited by in RCA: 25] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 127. | Ni YA, Chen H, Nie H, Zheng B, Gong Q. HMGB1: An overview of its roles in the pathogenesis of liver disease. J Leukoc Biol. 2021;110:987-998. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 4] [Cited by in RCA: 39] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
| 128. | Bansal R, van Baarlen J, Storm G, Prakash J. The interplay of the Notch signaling in hepatic stellate cells and macrophages determines the fate of liver fibrogenesis. Sci Rep. 2015;5:18272. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 46] [Cited by in RCA: 79] [Article Influence: 7.9] [Reference Citation Analysis (0)] |
| 129. | Martinez Lyons A, Boulter L. NOTCH signalling - a core regulator of bile duct disease? Dis Model Mech. 2023;16:dmm050231. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
| 130. | Sorrentino G. Microenvironmental control of the ductular reaction: balancing repair and disease progression. Cell Death Dis. 2025;16:246. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 5] [Article Influence: 5.0] [Reference Citation Analysis (0)] |
| 131. | Machado MV, Michelotti GA, Pereira TA, Xie G, Premont R, Cortez-Pinto H, Diehl AM. Accumulation of duct cells with activated YAP parallels fibrosis progression in non-alcoholic fatty liver disease. J Hepatol. 2015;63:962-970. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 94] [Cited by in RCA: 107] [Article Influence: 10.7] [Reference Citation Analysis (0)] |
| 132. | Pepe-Mooney BJ, Dill MT, Alemany A, Ordovas-Montanes J, Matsushita Y, Rao A, Sen A, Miyazaki M, Anakk S, Dawson PA, Ono N, Shalek AK, van Oudenaarden A, Camargo FD. Single-Cell Analysis of the Liver Epithelium Reveals Dynamic Heterogeneity and an Essential Role for YAP in Homeostasis and Regeneration. Cell Stem Cell. 2019;25:23-38.e8. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 116] [Cited by in RCA: 192] [Article Influence: 32.0] [Reference Citation Analysis (0)] |
| 133. | Verboven E, Moya IM, Sansores-Garcia L, Xie J, Hillen H, Kowalczyk W, Vella G, Verhulst S, Castaldo SA, Algueró-Nadal A, Romanelli L, Mercader-Celma C, Souza NA, Soheily S, Van Huffel L, Van Brussel T, Lambrechts D, Roskams T, Lemaigre FP, Bergers G, van Grunsven LA, Halder G. Regeneration Defects in Yap and Taz Mutant Mouse Livers Are Caused by Bile Duct Disruption and Cholestasis. Gastroenterology. 2021;160:847-862. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 20] [Cited by in RCA: 52] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
| 134. | Mavila N, Siraganahalli Eshwaraiah M, Kennedy J. Ductular Reactions in Liver Injury, Regeneration, and Disease Progression-An Overview. Cells. 2024;13:579. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 10] [Reference Citation Analysis (0)] |
| 135. | Rainu SK, Singh N. Miniaturized Liver Disease Mimics to Gain Insights into MMP Expression during Disease Progression. ACS Biomater Sci Eng. 2025;11:476-484. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 2] [Reference Citation Analysis (0)] |
| 136. | Sabir U, Gu HM, Zhang DW. Extracellular matrix turnover: phytochemicals target and modulate the dual role of matrix metalloproteinases (MMPs) in liver fibrosis. Phytother Res. 2023;37:4932-4962. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 14] [Reference Citation Analysis (0)] |
| 137. | Zhao X, Chen J, Sun H, Zhang Y, Zou D. New insights into fibrosis from the ECM degradation perspective: the macrophage-MMP-ECM interaction. Cell Biosci. 2022;12:117. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 48] [Cited by in RCA: 105] [Article Influence: 35.0] [Reference Citation Analysis (0)] |
| 138. | Zhang Z, Yang H, Han F, Guo P. Reactive Oxygen Species as Key Molecules in the Pathogenesis of Alcoholic Fatty Liver Disease and Nonalcoholic Fatty Liver Disease: Future Perspectives. Curr Issues Mol Biol. 2025;47:464. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 4] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 139. | Iracheta-Vellve A, Petrasek J, Gyogyosi B, Bala S, Csak T, Kodys K, Szabo G. Interleukin-1 inhibition facilitates recovery from liver injury and promotes regeneration of hepatocytes in alcoholic hepatitis in mice. Liver Int. 2017;37:968-973. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 26] [Cited by in RCA: 54] [Article Influence: 6.8] [Reference Citation Analysis (0)] |
| 140. | Zhou H, Liu Y, Su Y, Ji P, Kong L, Sun R, Zhang D, Xu H, Li W, Li W. Ginsenoside Rg1 attenuates lipopolysaccharide-induced chronic liver damage by activating Nrf2 signaling and inhibiting inflammasomes in hepatic cells. J Ethnopharmacol. 2024;324:117794. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 18] [Reference Citation Analysis (0)] |
| 141. | Varela-Rey M, Fontán-Gabás L, Blanco P, López-Zabalza MJ, Iraburu MJ. Glutathione depletion is involved in the inhibition of procollagen alpha1(I) mRNA levels caused by TNF-alpha on hepatic stellate cells. Cytokine. 2007;37:212-217. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 11] [Cited by in RCA: 18] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
| 142. | Kharaziha P, Hellström PM, Noorinayer B, Farzaneh F, Aghajani K, Jafari F, Telkabadi M, Atashi A, Honardoost M, Zali MR, Soleimani M. Improvement of liver function in liver cirrhosis patients after autologous mesenchymal stem cell injection: a phase I-II clinical trial. Eur J Gastroenterol Hepatol. 2009;21:1199-1205. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 317] [Cited by in RCA: 326] [Article Influence: 20.4] [Reference Citation Analysis (0)] |
| 143. | Jang YO, Kim YJ, Baik SK, Kim MY, Eom YW, Cho MY, Park HJ, Park SY, Kim BR, Kim JW, Soo Kim H, Kwon SO, Choi EH, Kim YM. Histological improvement following administration of autologous bone marrow-derived mesenchymal stem cells for alcoholic cirrhosis: a pilot study. Liver Int. 2014;34:33-41. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 126] [Cited by in RCA: 136] [Article Influence: 12.4] [Reference Citation Analysis (0)] |
| 144. | di Bonzo LV, Ferrero I, Cravanzola C, Mareschi K, Rustichell D, Novo E, Sanavio F, Cannito S, Zamara E, Bertero M, Davit A, Francica S, Novelli F, Colombatto S, Fagioli F, Parola M. Human mesenchymal stem cells as a two-edged sword in hepatic regenerative medicine: engraftment and hepatocyte differentiation versus profibrogenic potential. Gut. 2008;57:223-231. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 210] [Cited by in RCA: 218] [Article Influence: 12.8] [Reference Citation Analysis (0)] |
| 145. | Baertschiger RM, Serre-Beinier V, Morel P, Bosco D, Peyrou M, Clément S, Sgroi A, Kaelin A, Buhler LH, Gonelle-Gispert C. Fibrogenic potential of human multipotent mesenchymal stromal cells in injured liver. PLoS One. 2009;4:e6657. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 86] [Cited by in RCA: 100] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 146. | Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell. 2005;120:15-20. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8847] [Cited by in RCA: 9322] [Article Influence: 466.1] [Reference Citation Analysis (0)] |
| 147. | Kou M, Huang L, Yang J, Chiang Z, Chen S, Liu J, Guo L, Zhang X, Zhou X, Xu X, Yan X, Wang Y, Zhang J, Xu A, Tse HF, Lian Q. Mesenchymal stem cell-derived extracellular vesicles for immunomodulation and regeneration: a next generation therapeutic tool? Cell Death Dis. 2022;13:580. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 296] [Cited by in RCA: 340] [Article Influence: 113.3] [Reference Citation Analysis (0)] |
| 148. | Kim J, Lee C, Shin Y, Wang S, Han J, Kim M, Kim JM, Shin SC, Lee BJ, Kim TJ, Jung Y. sEVs from tonsil-derived mesenchymal stromal cells alleviate activation of hepatic stellate cells and liver fibrosis through miR-486-5p. Mol Ther. 2021;29:1471-1486. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 51] [Cited by in RCA: 50] [Article Influence: 12.5] [Reference Citation Analysis (0)] |
| 149. | Guy R, Offen D. Promising Opportunities for Treating Neurodegenerative Diseases with Mesenchymal Stem Cell-Derived Exosomes. Biomolecules. 2020;10:1320. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 43] [Cited by in RCA: 63] [Article Influence: 12.6] [Reference Citation Analysis (0)] |
| 150. | Chen J, Huang T, Liu R, Wang C, Jiang H, Sun H. Congenital microtia patients: the genetically engineered exosomes released from porous gelatin methacryloyl hydrogel for downstream small RNA profiling, functional modulation of microtia chondrocytes and tissue-engineered ear cartilage regeneration. J Nanobiotechnology. 2022;20:164. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 19] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
| 151. | Lou G, Chen L, Xia C, Wang W, Qi J, Li A, Zhao L, Chen Z, Zheng M, Liu Y. MiR-199a-modified exosomes from adipose tissue-derived mesenchymal stem cells improve hepatocellular carcinoma chemosensitivity through mTOR pathway. J Exp Clin Cancer Res. 2020;39:4. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 96] [Cited by in RCA: 221] [Article Influence: 44.2] [Reference Citation Analysis (0)] |
| 152. | Hyun J, Wang S, Kim J, Kim GJ, Jung Y. MicroRNA125b-mediated Hedgehog signaling influences liver regeneration by chorionic plate-derived mesenchymal stem cells. Sci Rep. 2015;5:14135. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 111] [Cited by in RCA: 115] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
| 153. | Li J, Ghazwani M, Zhang Y, Lu J, Li J, Fan J, Gandhi CR, Li S. miR-122 regulates collagen production via targeting hepatic stellate cells and suppressing P4HA1 expression. J Hepatol. 2013;58:522-528. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 139] [Cited by in RCA: 161] [Article Influence: 13.4] [Reference Citation Analysis (0)] |
| 154. | Lou G, Yang Y, Liu F, Ye B, Chen Z, Zheng M, Liu Y. MiR-122 modification enhances the therapeutic efficacy of adipose tissue-derived mesenchymal stem cells against liver fibrosis. J Cell Mol Med. 2017;21:2963-2973. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 107] [Cited by in RCA: 176] [Article Influence: 22.0] [Reference Citation Analysis (0)] |
| 155. | Matsumoto Y, Itami S, Kuroda M, Yoshizato K, Kawada N, Murakami Y. MiR-29a Assists in Preventing the Activation of Human Stellate Cells and Promotes Recovery From Liver Fibrosis in Mice. Mol Ther. 2016;24:1848-1859. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 46] [Cited by in RCA: 73] [Article Influence: 8.1] [Reference Citation Analysis (0)] |
| 156. | Kwiecinski M, Elfimova N, Noetel A, Töx U, Steffen HM, Hacker U, Nischt R, Dienes HP, Odenthal M. Expression of platelet-derived growth factor-C and insulin-like growth factor I in hepatic stellate cells is inhibited by miR-29. Lab Invest. 2012;92:978-987. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 65] [Cited by in RCA: 69] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
| 157. | Ji J, Zhao L, Budhu A, Forgues M, Jia HL, Qin LX, Ye QH, Yu J, Shi X, Tang ZY, Wang XW. Let-7g targets collagen type I alpha2 and inhibits cell migration in hepatocellular carcinoma. J Hepatol. 2010;52:690-697. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 166] [Cited by in RCA: 175] [Article Influence: 11.7] [Reference Citation Analysis (0)] |
| 158. | Feng GX, Li J, Yang Z, Zhang SQ, Liu YX, Zhang WY, Ye LH, Zhang XD. Hepatitis B virus X protein promotes the development of liver fibrosis and hepatoma through downregulation of miR-30e targeting P4HA2 mRNA. Oncogene. 2017;36:6895-6905. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 32] [Cited by in RCA: 49] [Article Influence: 6.1] [Reference Citation Analysis (0)] |
| 159. | Wang S, Huang M, Wang Z, Wang W, Zhang Z, Qu S, Liu C. MicroRNA133b targets TGFβ receptor I to inhibit TGFβinduced epithelialtomesenchymal transition and metastasis by suppressing the TGFβ/SMAD pathway in breast cancer. Int J Oncol. 2019;55:1097-1109. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 3] [Cited by in RCA: 14] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
| 160. | Lakner AM, Steuerwald NM, Walling TL, Ghosh S, Li T, McKillop IH, Russo MW, Bonkovsky HL, Schrum LW. Inhibitory effects of microRNA 19b in hepatic stellate cell-mediated fibrogenesis. Hepatology. 2012;56:300-310. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 152] [Cited by in RCA: 173] [Article Influence: 13.3] [Reference Citation Analysis (0)] |
| 161. | Xiong M, Jiang L, Zhou Y, Qiu W, Fang L, Tan R, Wen P, Yang J. The miR-200 family regulates TGF-β1-induced renal tubular epithelial to mesenchymal transition through Smad pathway by targeting ZEB1 and ZEB2 expression. Am J Physiol Renal Physiol. 2012;302:F369-F379. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 192] [Cited by in RCA: 208] [Article Influence: 14.9] [Reference Citation Analysis (1)] |
| 162. | Roderburg C, Urban GW, Bettermann K, Vucur M, Zimmermann H, Schmidt S, Janssen J, Koppe C, Knolle P, Castoldi M, Tacke F, Trautwein C, Luedde T. Micro-RNA profiling reveals a role for miR-29 in human and murine liver fibrosis. Hepatology. 2011;53:209-218. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 656] [Cited by in RCA: 664] [Article Influence: 47.4] [Reference Citation Analysis (0)] |
| 163. | Choi DW, Cho KA, Kim J, Lee HJ, Kim YH, Park JW, Woo SY. Extracellular vesicles from tonsilderived mesenchymal stromal cells show antitumor effect via miR199a3p. Int J Mol Med. 2021;48:221. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 4] [Cited by in RCA: 12] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 164. | Aung SW, Abu Kasim NH, Shamsuddin SAA, Ramasamy TS. MicroRNAomic Transcriptomic Analysis Reveal Deregulation of Clustered Cellular Functions in Human Mesenchymal Stem Cells During in Vitro Passaging. Stem Cell Rev Rep. 2020;16:222-238. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 2] [Cited by in RCA: 5] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
| 165. | Zheng M, Li YY, Wang GF, Jin JY, Wang YH, Wang TM, Yang L, Liu SY, Wu JS, Wang ZT, Ma YM. Protective effect of cultured bear bile powder against dimethylnitrosamine-induced hepatic fibrosis in rats. Biomed Pharmacother. 2019;112:108701. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 19] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
| 166. | Feng M, Ding J, Wang M, Zhang J, Zhu X, Guan W. Kupffer-derived matrix metalloproteinase-9 contributes to liver fibrosis resolution. Int J Biol Sci. 2018;14:1033-1040. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 30] [Cited by in RCA: 78] [Article Influence: 11.1] [Reference Citation Analysis (0)] |
| 167. | Caligiuri A, Gentilini A, Pastore M, Gitto S, Marra F. Cellular and Molecular Mechanisms Underlying Liver Fibrosis Regression. Cells. 2021;10:2759. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 15] [Cited by in RCA: 180] [Article Influence: 45.0] [Reference Citation Analysis (0)] |
| 168. | Cordero-Espinoza L, Huch M. The balancing act of the liver: tissue regeneration versus fibrosis. J Clin Invest. 2018;128:85-96. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 162] [Cited by in RCA: 168] [Article Influence: 24.0] [Reference Citation Analysis (0)] |
| 169. | Krenkel O, Puengel T, Govaere O, Abdallah AT, Mossanen JC, Kohlhepp M, Liepelt A, Lefebvre E, Luedde T, Hellerbrand C, Weiskirchen R, Longerich T, Costa IG, Anstee QM, Trautwein C, Tacke F. Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis. Hepatology. 2018;67:1270-1283. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 290] [Cited by in RCA: 424] [Article Influence: 60.6] [Reference Citation Analysis (0)] |
| 170. | Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen MJ, Manon-Jensen T, Siebuhr A, Gudmann NS, Rønnow S, Sand JM, Daniels SJ, Mortensen JH, Schuppan D. The good and the bad collagens of fibrosis - Their role in signaling and organ function. Adv Drug Deliv Rev. 2017;121:43-56. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 227] [Cited by in RCA: 371] [Article Influence: 46.4] [Reference Citation Analysis (1)] |
| 171. | Li J, Guo C, Wu J. Astaxanthin in Liver Health and Disease: A Potential Therapeutic Agent. Drug Des Devel Ther. 2020;14:2275-2285. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 19] [Cited by in RCA: 49] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
| 172. | Miao Y, Wu Y, Jin Y, Lei M, Nan J, Wu X. Benzoquinone derivatives with antioxidant activity inhibit activated hepatic stellate cells and attenuate liver fibrosis in TAA-induced mice. Chem Biol Interact. 2020;317:108945. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 20] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
| 173. | Lee YA, Wallace MC, Friedman SL. Pathobiology of liver fibrosis: a translational success story. Gut. 2015;64:830-841. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 601] [Cited by in RCA: 713] [Article Influence: 71.3] [Reference Citation Analysis (0)] |
| 174. | Issa R, Williams E, Trim N, Kendall T, Arthur MJ, Reichen J, Benyon RC, Iredale JP. Apoptosis of hepatic stellate cells: involvement in resolution of biliary fibrosis and regulation by soluble growth factors. Gut. 2001;48:548-557. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 235] [Cited by in RCA: 248] [Article Influence: 10.3] [Reference Citation Analysis (0)] |
| 175. | El Taghdouini A, Najimi M, Sancho-Bru P, Sokal E, van Grunsven LA. In vitro reversion of activated primary human hepatic stellate cells. Fibrogenesis Tissue Repair. 2015;8:14. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 48] [Cited by in RCA: 76] [Article Influence: 7.6] [Reference Citation Analysis (0)] |
| 176. | Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, Iwaisako K, Moore-Morris T, Scott B, Tsukamoto H, Evans SM, Dillmann W, Glass CK, Brenner DA. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A. 2012;109:9448-9453. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 507] [Cited by in RCA: 643] [Article Influence: 49.5] [Reference Citation Analysis (0)] |
| 177. | Nishizawa H, Iguchi G, Fukuoka H, Takahashi M, Suda K, Bando H, Matsumoto R, Yoshida K, Odake Y, Ogawa W, Takahashi Y. IGF-I induces senescence of hepatic stellate cells and limits fibrosis in a p53-dependent manner. Sci Rep. 2016;6:34605. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 78] [Cited by in RCA: 126] [Article Influence: 14.0] [Reference Citation Analysis (0)] |
| 178. | Tan S, Liu H, Ke B, Jiang J, Wu B. The peripheral CB(1) receptor antagonist JD5037 attenuates liver fibrosis via a CB(1) receptor/β-arrestin1/Akt pathway. Br J Pharmacol. 2020;177:2830-2847. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 10] [Cited by in RCA: 26] [Article Influence: 5.2] [Reference Citation Analysis (0)] |
| 179. | Campana L, Iredale JP. Regression of Liver Fibrosis. Semin Liver Dis. 2017;37:1-10. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 214] [Cited by in RCA: 290] [Article Influence: 36.3] [Reference Citation Analysis (0)] |
| 180. | Zhang Y, Hua L, Lin C, Yuan M, Xu W, Raj D A, Venkidasamy B, Cespedes-Acuna CL, Nile SH, Yan G, Zheng H. Pien-Tze-Huang alleviates CCl(4)-induced liver fibrosis through the inhibition of HSC autophagy and the TGF-β1/Smad2 pathway. Front Pharmacol. 2022;13:937484. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 15] [Reference Citation Analysis (0)] |
| 181. | Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev. 2017;121:27-42. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 530] [Cited by in RCA: 1103] [Article Influence: 137.9] [Reference Citation Analysis (0)] |
| 182. | Kong Z, Liu R, Cheng Y. Artesunate alleviates liver fibrosis by regulating ferroptosis signaling pathway. Biomed Pharmacother. 2019;109:2043-2053. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 124] [Cited by in RCA: 282] [Article Influence: 40.3] [Reference Citation Analysis (0)] |
| 183. | Gonzalez SA, Fiel MI, Sauk J, Canchis PW, Liu RC, Chiriboga L, Yee HT, Jacobson IM, Talal AH. Inverse association between hepatic stellate cell apoptosis and fibrosis in chronic hepatitis C virus infection. J Viral Hepat. 2009;16:141-148. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 22] [Cited by in RCA: 26] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
| 184. | Xu T, Ni MM, Xing-Li, Li XF, Meng XM, Huang C, Li J. NLRC5 regulates TGF-β1-induced proliferation and activation of hepatic stellate cells during hepatic fibrosis. Int J Biochem Cell Biol. 2016;70:92-104. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 34] [Cited by in RCA: 46] [Article Influence: 4.6] [Reference Citation Analysis (0)] |
| 185. | Koda Y, Teratani T, Chu PS, Hagihara Y, Mikami Y, Harada Y, Tsujikawa H, Miyamoto K, Suzuki T, Taniki N, Sujino T, Sakamoto M, Kanai T, Nakamoto N. CD8(+) tissue-resident memory T cells promote liver fibrosis resolution by inducing apoptosis of hepatic stellate cells. Nat Commun. 2021;12:4474. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 22] [Cited by in RCA: 145] [Article Influence: 36.3] [Reference Citation Analysis (0)] |
| 186. | Liu H, Dai L, Wang M, Feng F, Xiao Y. Tunicamycin Induces Hepatic Stellate Cell Apoptosis Through Calpain-2/Ca(2 +)-Dependent Endoplasmic Reticulum Stress Pathway. Front Cell Dev Biol. 2021;9:684857. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 2] [Cited by in RCA: 9] [Article Influence: 2.3] [Reference Citation Analysis (0)] |
| 187. | Jelski W, Szmitkowski M. Alcohol dehydrogenase (ADH) and aldehyde dehydrogenase (ALDH) in the cancer diseases. Clin Chim Acta. 2008;395:1-5. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 102] [Cited by in RCA: 122] [Article Influence: 7.2] [Reference Citation Analysis (0)] |
| 188. | Gao N, Li J, Li MR, Qi B, Wang Z, Wang GJ, Gao J, Qiao HL. Higher Activity of Alcohol Dehydrogenase Is Correlated with Hepatic Fibrogenesis. J Pharmacol Exp Ther. 2018;367:473-482. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 9] [Cited by in RCA: 11] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
| 189. | Liu X, Xu J, Rosenthal S, Zhang LJ, McCubbin R, Meshgin N, Shang L, Koyama Y, Ma HY, Sharma S, Heinz S, Glass CK, Benner C, Brenner DA, Kisseleva T. Identification of Lineage-Specific Transcription Factors That Prevent Activation of Hepatic Stellate Cells and Promote Fibrosis Resolution. Gastroenterology. 2020;158:1728-1744.e14. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 104] [Cited by in RCA: 140] [Article Influence: 28.0] [Reference Citation Analysis (0)] |
| 190. | Troeger JS, Mederacke I, Gwak GY, Dapito DH, Mu X, Hsu CC, Pradere JP, Friedman RA, Schwabe RF. Deactivation of hepatic stellate cells during liver fibrosis resolution in mice. Gastroenterology. 2012;143:1073-83.e22. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 380] [Cited by in RCA: 404] [Article Influence: 31.1] [Reference Citation Analysis (0)] |
| 191. | Iredale JP, Benyon RC, Pickering J, McCullen M, Northrop M, Pawley S, Hovell C, Arthur MJ. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102:538-549. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 785] [Cited by in RCA: 834] [Article Influence: 30.9] [Reference Citation Analysis (0)] |
| 192. | Song G, Pacher M, Balakrishnan A, Yuan Q, Tsay HC, Yang D, Reetz J, Brandes S, Dai Z, Pützer BM, Araúzo-Bravo MJ, Steinemann D, Luedde T, Schwabe RF, Manns MP, Schöler HR, Schambach A, Cantz T, Ott M, Sharma AD. Direct Reprogramming of Hepatic Myofibroblasts into Hepatocytes In Vivo Attenuates Liver Fibrosis. Cell Stem Cell. 2016;18:797-808. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 138] [Cited by in RCA: 170] [Article Influence: 18.9] [Reference Citation Analysis (0)] |
| 193. | Kim N, Choi S, Lim C, Lee H, Oh J. Albumin mediates PPAR-gamma or C/EBP-alpha-induced phenotypic changes in pancreatic stellate cells. Biochem Biophys Res Commun. 2010;391:640-644. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 14] [Cited by in RCA: 20] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
| 194. | Miyahara T, Schrum L, Rippe R, Xiong S, Yee HF Jr, Motomura K, Anania FA, Willson TM, Tsukamoto H. Peroxisome proliferator-activated receptors and hepatic stellate cell activation. J Biol Chem. 2000;275:35715-35722. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 362] [Cited by in RCA: 377] [Article Influence: 15.1] [Reference Citation Analysis (0)] |
| 195. | Hazra S, Xiong S, Wang J, Rippe RA, Krishna V, Chatterjee K, Tsukamoto H. Peroxisome proliferator-activated receptor gamma induces a phenotypic switch from activated to quiescent hepatic stellate cells. J Biol Chem. 2004;279:11392-11401. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 236] [Cited by in RCA: 255] [Article Influence: 11.6] [Reference Citation Analysis (0)] |
| 196. | Nakano Y, Kamiya A, Sumiyoshi H, Tsuruya K, Kagawa T, Inagaki Y. A Deactivation Factor of Fibrogenic Hepatic Stellate Cells Induces Regression of Liver Fibrosis in Mice. Hepatology. 2020;71:1437-1452. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 61] [Cited by in RCA: 68] [Article Influence: 13.6] [Reference Citation Analysis (0)] |
| 197. | Jiménez Calvente C, Sehgal A, Popov Y, Kim YO, Zevallos V, Sahin U, Diken M, Schuppan D. Specific hepatic delivery of procollagen α1(I) small interfering RNA in lipid-like nanoparticles resolves liver fibrosis. Hepatology. 2015;62:1285-1297. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 83] [Cited by in RCA: 108] [Article Influence: 10.8] [Reference Citation Analysis (0)] |
| 198. | Wang YH, Suk FM, Liu CL, Chen TL, Twu YC, Hsu MH, Liao YJ. Antifibrotic Effects of a Barbituric Acid Derivative on Liver Fibrosis by Blocking the NF-κB Signaling Pathway in Hepatic Stellate Cells. Front Pharmacol. 2020;11:388. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 8] [Cited by in RCA: 15] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
| 199. | Xu F, Liu C, Zhou D, Zhang L. TGF-β/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J Histochem Cytochem. 2016;64:157-167. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 326] [Cited by in RCA: 590] [Article Influence: 65.6] [Reference Citation Analysis (0)] |
| 200. | Nishikawa K, Osawa Y, Kimura K. Wnt/β-Catenin Signaling as a Potential Target for the Treatment of Liver Cirrhosis Using Antifibrotic Drugs. Int J Mol Sci. 2018;19:3103. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 49] [Cited by in RCA: 92] [Article Influence: 13.1] [Reference Citation Analysis (0)] |
| 201. | Xie G, Karaca G, Swiderska-Syn M, Michelotti GA, Krüger L, Chen Y, Premont RT, Choi SS, Diehl AM. Cross-talk between Notch and Hedgehog regulates hepatic stellate cell fate in mice. Hepatology. 2013;58:1801-1813. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 86] [Cited by in RCA: 107] [Article Influence: 8.9] [Reference Citation Analysis (0)] |
| 202. | Mannaerts I, Leite SB, Verhulst S, Claerhout S, Eysackers N, Thoen LF, Hoorens A, Reynaert H, Halder G, van Grunsven LA. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J Hepatol. 2015;63:679-688. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 230] [Cited by in RCA: 321] [Article Influence: 32.1] [Reference Citation Analysis (1)] |
| 203. | Tao Y, Wang N, Qiu T, Sun X. The Role of Autophagy and NLRP3 Inflammasome in Liver Fibrosis. Biomed Res Int. 2020;2020:7269150. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 13] [Cited by in RCA: 48] [Article Influence: 9.6] [Reference Citation Analysis (0)] |
| 204. | Song YR, Jang MH, Jang B, Bae SJ, Bak SB, Lee SM, Yun U, Lee JH, Park SM, Jung DH, Sa BS, Song JK, Lee EH, Kim KY, Park K, Kim YW, Kim SC. Jageum-Jung, the herbal pharmaceuticals, inhibits the hepatic fibrogenesis as mediated with TGF-β1/smad signaling. Mol Cell Toxicol. 2022;18:243-251. [RCA] [DOI] [Full Text] [Cited by in RCA: 4] [Reference Citation Analysis (0)] |
| 205. | Dewidar B, Soukupova J, Fabregat I, Dooley S. TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis: Updated. Curr Pathobiol Rep. 2015;3:291-305. [RCA] [DOI] [Full Text] [Cited by in Crossref: 23] [Cited by in RCA: 35] [Article Influence: 3.5] [Reference Citation Analysis (0)] |
| 206. | Hou J, Tian J, Jiang W, Gao Y, Fu F. Therapeutic effects of SMND-309, a new metabolite of salvianolic acid B, on experimental liver fibrosis. Eur J Pharmacol. 2011;650:390-395. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 18] [Cited by in RCA: 21] [Article Influence: 1.4] [Reference Citation Analysis (0)] |
| 207. | Thakral S, Ghoshal K. miR-122 is a unique molecule with great potential in diagnosis, prognosis of liver disease, and therapy both as miRNA mimic and antimir. Curr Gene Ther. 2015;15:142-150. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 136] [Cited by in RCA: 186] [Article Influence: 18.6] [Reference Citation Analysis (0)] |
| 208. | Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, Persson R, King BD, Kauppinen S, Levin AA, Hodges MR. Treatment of HCV infection by targeting microRNA. N Engl J Med. 2013;368:1685-1694. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 1665] [Cited by in RCA: 1717] [Article Influence: 143.1] [Reference Citation Analysis (0)] |
| 209. | Krützfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with 'antagomirs'. Nature. 2005;438:685-689. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 3005] [Cited by in RCA: 3087] [Article Influence: 154.4] [Reference Citation Analysis (0)] |
| 210. | Wang YZ, Zhang W, Wang YH, Fu XL, Xue CQ. Repression of liver cirrhosis achieved by inhibitory effect of miR-454 on hepatic stellate cells activation and proliferation via Wnt10a. J Biochem. 2019;165:361-367. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 13] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
| 211. | Chen Y, Ou Y, Dong J, Yang G, Zeng Z, Liu Y, Liu B, Li W, He X, Lan T. Osteopontin promotes collagen I synthesis in hepatic stellate cells by miRNA-129-5p inhibition. Exp Cell Res. 2018;362:343-348. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 31] [Cited by in RCA: 42] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
| 212. | Cheng K, Mahato RI. Biological and therapeutic applications of small RNAs. Pharm Res. 2011;28:2961-2965. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 8] [Cited by in RCA: 13] [Article Influence: 0.9] [Reference Citation Analysis (0)] |
| 213. | Li C, Nguyen HT, Zhuang Y, Lin Y, Flemington EK, Guo W, Guenther J, Burow ME, Morris GF, Sullivan D, Shan B. Post-transcriptional up-regulation of miR-21 by type I collagen. Mol Carcinog. 2011;50:563-570. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 26] [Cited by in RCA: 27] [Article Influence: 1.9] [Reference Citation Analysis (0)] |
| 214. | Shi R, Gao S, Zhang J, Xu J, Graham LM, Yang X, Li C. Collagen prolyl 4-hydroxylases modify tumor progression. Acta Biochim Biophys Sin (Shanghai). 2021;53:805-814. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 25] [Cited by in RCA: 39] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
| 215. | Ichimura A, Ruike Y, Terasawa K, Tsujimoto G. miRNAs and regulation of cell signaling. FEBS J. 2011;278:1610-1618. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 63] [Cited by in RCA: 63] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
| 216. | Inui M, Martello G, Piccolo S. MicroRNA control of signal transduction. Nat Rev Mol Cell Biol. 2010;11:252-263. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 958] [Cited by in RCA: 930] [Article Influence: 62.0] [Reference Citation Analysis (0)] |
| 217. | Spizzo R, Rushworth D, Guerrero M, Calin GA. RNA inhibition, microRNAs, and new therapeutic agents for cancer treatment. Clin Lymphoma Myeloma. 2009;9 Suppl 3:S313-S318. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 26] [Cited by in RCA: 24] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
| 218. | Bala S, Marcos M, Kodys K, Csak T, Catalano D, Mandrekar P, Szabo G. Up-regulation of microRNA-155 in macrophages contributes to increased tumor necrosis factor {alpha} (TNF{alpha}) production via increased mRNA half-life in alcoholic liver disease. J Biol Chem. 2011;286:1436-1444. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 301] [Cited by in RCA: 339] [Article Influence: 22.6] [Reference Citation Analysis (0)] |
| 219. | Bala S, Marcos M, Szabo G. Emerging role of microRNAs in liver diseases. World J Gastroenterol. 2009;15:5633-5640. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in CrossRef: 114] [Cited by in RCA: 122] [Article Influence: 7.6] [Reference Citation Analysis (0)] |
| 220. | Bala S, Szabo G. MicroRNA Signature in Alcoholic Liver Disease. Int J Hepatol. 2012;2012:498232. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 87] [Cited by in RCA: 107] [Article Influence: 8.2] [Reference Citation Analysis (0)] |
| 221. | Lan HY, Chung ACK. Transforming growth factor-β and Smads. Contrib Nephrol. 2011;170:75-82. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 80] [Cited by in RCA: 91] [Article Influence: 6.5] [Reference Citation Analysis (0)] |
| 222. | Zou Y, Li S, Li Z, Song D, Zhang S, Yao Q. MiR-146a attenuates liver fibrosis by inhibiting transforming growth factor-β1 mediated epithelial-mesenchymal transition in hepatocytes. Cell Signal. 2019;58:1-8. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 18] [Cited by in RCA: 37] [Article Influence: 6.2] [Reference Citation Analysis (0)] |
| 223. | Chen X, Xiao W, Chen W, Liu X, Wu M, Bo Q, Luo Y, Ye S, Cao Y, Liu Y. MicroRNA-26a and -26b inhibit lens fibrosis and cataract by negatively regulating Jagged-1/Notch signaling pathway. Cell Death Differ. 2017;24:1431-1442. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 40] [Cited by in RCA: 73] [Article Influence: 9.1] [Reference Citation Analysis (0)] |
| 224. | Wen D, Zhang H, Zhou Y, Wang J. The Molecular Mechanisms and Function of miR-15a/16 Dysregulation in Fibrotic Diseases. Int J Mol Sci. 2022;23:16041. [RCA] [PubMed] [DOI] [Full Text] [Cited by in RCA: 8] [Reference Citation Analysis (0)] |
| 225. | Zhang L, Qiu Y, Yang F, Yao J, Wang Y, Qin Y, Mou H, Jing Q, Liu L, Ju Z. Hepatic microRNA-126 deficiency restrains liver regeneration through p53 pathway in mice. Signal Transduct Target Ther. 2021;6:32. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in RCA: 5] [Reference Citation Analysis (0)] |
| 226. | Wang X, Seo W, Park SH, Fu Y, Hwang S, Rodrigues RM, Feng D, Gao B, He Y. MicroRNA-223 restricts liver fibrosis by inhibiting the TAZ-IHH-GLI2 and PDGF signaling pathways via the crosstalk of multiple liver cell types. Int J Biol Sci. 2021;17:1153-1167. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 17] [Cited by in RCA: 32] [Article Influence: 8.0] [Reference Citation Analysis (0)] |
| 227. | Ye D, Zhang T, Lou G, Liu Y. Role of miR-223 in the pathophysiology of liver diseases. Exp Mol Med. 2018;50:1-12. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 52] [Cited by in RCA: 91] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
| 228. | Gu J, Xu H, Chen Y, Li N, Hou X. MiR-223 as a Regulator and Therapeutic Target in Liver Diseases. Front Immunol. 2022;13:860661. [RCA] [PubMed] [DOI] [Full Text] [Full Text (PDF)] [Cited by in Crossref: 1] [Cited by in RCA: 31] [Article Influence: 10.3] [Reference Citation Analysis (0)] |
| 229. | Chen W, Yan X, Yang A, Xu A, Huang T, You H. miRNA-150-5p promotes hepatic stellate cell proliferation and sensitizes hepatocyte apoptosis during liver fibrosis. Epigenomics. 2020;12:53-67. [RCA] [PubMed] [DOI] [Full Text] [Cited by in Crossref: 13] [Cited by in RCA: 27] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
