Skip to main content

Targeting HSP47 and HSP70: promising therapeutic approaches in liver fibrosis management

Abstract

Liver fibrosis is a liver disease in which there is an excessive buildup of extracellular matrix proteins, including collagen. By regulating cytokine production and the inflammatory response, heat shock proteins (HSPs) contribute significantly to a wider spectrum of fibrotic illnesses, such as lung, liver, and idiopathic pulmonary fibrosis by aiding in the folding and assembly of freshly synthesized proteins, HSPs serve as chaperones. HSP70 is one of the key HSPs in avoiding protein aggregation which induces its action by sending unfolded and/or misfolded proteins to the ubiquitin–proteasome degradation pathway and antagonizing influence on epithelial-mesenchymal transition. HSP47, on the other hand, is crucial for boosting collagen synthesis, and deposition, and fostering the emergence of fibrotic disorders. The current review aims to provide light on how HSP70 and HSP47 affect hepatic fibrogenesis. Additionally, our review looks into new therapeutic approaches that target HSP70 and HSP47 and could potentially be used as drug candidates to treat liver fibrosis, especially in cases of comorbidities.

Introduction

Liver fibrosis is an inflammatory response brought on by a variety of conditions, including alcohol use, non-alcoholic steatohepatitis (NASH), viral hepatitis (hepatitis B (HBV) and hepatitis C (HCV)), autoimmune hepatitis, non-alcoholic fatty liver disease (NAFLD), and cholestatic liver diseases [1]. The formation of a chronic inflammatory response results in an aberrant wound healing response which induces extracellular matrix (ECM) components accumulation in the liver and thus the creation of fibrous scar tissue. The existence of a fibrous scar alters the architecture of the liver, leading to hepatocyte loss, the disruption of normal liver function, and ultimately liver failure [2, 3]. Unless it progresses and becomes cirrhosis, liver fibrosis can be reversed.

Liver diseases whatever result from pathogenic, toxic, metabolic, or viral causes induce hepatocyte damage and immune cell infiltration that activates the trans-differentiation of hepatic stellate cells (HSCs) into myofibroblasts that produce collagen [4]. HSCs differentiate into myofibroblasts, begin expressing alpha-smooth muscle actin (α-SMA), move to tissue healing sites, and secrete a considerable amount of ECM [5]. Myofibroblasts may undergo apoptosis and inactivation after the liver injury is removed [6].

Through a process known as epithelial-mesenchymal transition (EMT), epithelial cells that are normally found on the exterior of blood vessels and organs may lose their polarity, migrate, and give rise to myofibroblasts [6]. Interestingly, Xie and Diehl [7] found that extended culture of cholangiocytes and hepatocytes increases the expression of α-SMA while decreasing the expression of epithelial markers.

Transforming growth factor-beta 1 (TGF-β1) mediates the activation of portal fibrosis during cholestatic liver fibrosis, which includes the interaction of mesothelin with a MUC16-Thy1- TGF-β1RI complex [8, 9] and platelet-derived growth factor (PDGF), as well as increased contractility, and high levels of α-SMA, and connective tissue growth factor (CTGF) [10].

Myofibroblasts are physiologically implicated in tissue regeneration; however, after a short-term insult, anti-fibrotic mechanisms balance this activity, leading to myofibroblast inactivation or apoptosis and scar resolution. In contrast, chronic liver disorders result in prolonged activation of proliferative, contractile, and migratory myofibroblasts that result in an excess synthesis of ECM due to an imbalance of pro- and anti-fibrogenic pathways. ECM consists of type I and III collagen and fibronectin and its presence depends on the balance between matrix metalloproteinase (MMP) and tissue inhibitors of metalloproteinases (TIMP). ECM predominates as MMPs activity decreases and TIMPs activity increases [11].

AMPK (5' adenosine monophosphate-activated protein kinase) is a metabolic master regulator that regulates cellular energy homeostasis [12]. AMPK activity has been shown to suppress HSC activation by decreasing the activation of either nuclear factor kappa B (NF-kB) or mammalian target of rapamycin (mTOR) signaling [13]. In mice, adiponectin (an AMPK activator) deficiency exacerbated CCl4-induced fibrosis [14]. Furthermore, activation of AMPK in human HSCs by adiponectin or 5-Aminoimidazole-4-carboxamide ribonucleoside (AICAR) reduced HSC activation and migration in response to PDGF [13]. Likewise, inhibiting AMPK boosted PDGF-induced HSC proliferation, migration, and activation [13].

Non-parenchymal cells (NPCs), such as Kupffer cells and other immune cells, play a major role in determining whether the liver enters an anti-fibrotic scar-dissolving stage or advances into an unchecked fibrosis-promoting stage. Hepatocyte apoptosis and the release of damage-associated patterns (DAMPs) also cause the recruitment and activation of lymphocytes and macrophages, which promote HSC trans-differentiation and myofibroblast activation by producing pro-inflammatory and pro-fibrogenic cytokines to induce inflammation, such as PDGF, tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1β), as well as activating the TGF-β1/Smad signal pathway, mitogen-activated protein kinase (MAPK) [1518].

Additionally, by producing and secreting pro-inflammatory and pro-fibrogenic chemicals including apoptosis-signal-regulating kinase 1, pan-caspase, and galectin-3, monocytes further damage hepatocytes, encourage the activation of HSCs, and exacerbate inflammation and fibrosis. TGF-β1 also promotes the conversion of monocytes into macrophages which secrete inflammatory mediators like IL-1 and IL-6 that encourages the escalation of the inflammatory response and the ongoing activation and survival of HSCs [19].

Besides all these HSCs activators, there is a promising approach that activates HSCs through control of different heat shock proteins (HSPs) levels and is thus considered a new therapeutic target for the management of liver fibrosis.

HSPs and liver fibrosis

Heat shock proteins are stress proteins that cannot be activated under normal conditions because their expression is rigorously controlled by a variety of environmental and physiological insults, such as heat shock, oxidative stress, heavy metals, ultraviolet radiation, and membrane perturbations, either to aid in cell survival or to promote the death of an irreparably damaged cell [20, 21]. Based on their molecular weights, the HSPs family is divided into HSP100, HSP90, HSP70, HSP60, and HSP47. In fibrosis, HSPs are crucial for collagen formation in addition to their roles in anti-oxidation, synergistic immunity, and anti-apoptosis. HSPs may therefore be intimately linked to the development or the prevention of fibrogenesis and fibrosis [22]. Among the most prevalent HSPs that affect liver fibrogenesis are the pro-fibrotic HSP47 and the anti-fibrotic HSP70.

A-Role of HSP70 in the regulation of liver fibrosis

In response to numerous stimuli, such as heat, oxidative stress, and chemical damage, HSP70 is up-regulated in cells to help in avoiding protein aggregation [23]. Additionally, EMT, a player in the fibrosis process, is negatively impacted by HSP70 [24]. Sellares and Veraldi [25] mentioned that Hsp70 deficiency contributes to fibrosis, and interventions aimed at restoring normal Hsp70 expression represent a novel therapeutic strategy for fibrosis.

B-Role of HSP47 in the regulation of liver fibrosis

HSP47 is presented in the endoplasmic reticulum and is crucial in controlling collagen synthesis. HSP47 is implicated in fibrotic disorders such as scleroderma, renal interstitial fibrosis, peritoneal fibrosis, cardiac fibrosis, intestinal fibrosis, keloid fibrosis, and pulmonary fibrosis by encouraging the buildup of collagen [26].

Chronic hepatitis B and chronic schistosomiasis patients both had elevated levels of HSP47, TGF-β1, and CTGF. HSP47 mRNA expression considerably increased as schistosomiasis hepatic fibrosis progressed [27, 28] which makes HSP47 a biomarker for schistosomal hepatic fibrosis in its early stages [29, 30].

Additionally, HSP47-targeted small interfering RNA (siRNA) and short hairpin RNA (shRNA) can reduce collagen formation in mice with hepatic fibrosis brought on by Schistosoma japonicum [31].

The expression of HSP47, endothelin receptor A (ETAR), and endothelin receptor B (ETBR) was significantly increased in mice models of liver fibrosis caused by Schistosoma japonicum. When HSP47 shRNA was applied in vitro and in vivo, HSP47 expression was significantly reduced which decreased ETAR and ETBR levels on the cell membrane surface [32].

HSCs are the source of HSP47 expression and blocking HSCs activation can diminish the synthesis of HSP47, hence limiting or avoiding liver fibrosis [32]. HSP47 inhibition significantly suppressed collagen production in fibroblasts in vitro in the ulcerative colitis model [33]. HSP47 synthesis may be regulated by heat shock factor 1 (HSF-1) activation. Inactivation of HSF1 by both tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and siRNA results in the down-regulation of HSP47, lowering collagen buildup, and delaying the fibrosis process [34].

Here, we will discuss different regulators of Both HSP70 and HSP47 that aids in controlling liver fibrosis.

Factors regulate HSPs activity

TGF-β/Smad4 signaling pathway and HSPs

MiR-455-3p decreases HSF-1 expression and limits HSC activation by inhibiting the HSP47/TGF-β1/Smad4 signaling pathway [35]. Furthermore, miR-125b, miR-378, and miR-152 can prevent liver fibrosis by modulating GLI family zinc expression [36].

TGF-βIR can phosphorylate SMAD2 and SMAD3, which suggests that phosphorylation of SMAD2 and SMAD3, is required for the smooth transmission of the TGF-β signaling pathway and thus its activation and accelerating the development of liver fibrosis [37, 38]. Hsp70 can inhibit the phosphorylation of these two SMADs by interacting with SMAD2 and SMAD3, thereby inhibiting the conduction of the TGF-β signaling pathway [39].

Nuclear receptors (NRs) and HSPs

The removal of a stabilizing HSP aimed directly at gene transcription in the cell nucleus activates type-1 NRs before dimerization in the cytoplasm [40]. Hsp40-induced ATP hydrolysis delivers protein substrate to Hsp70 and increases Hsp70 association with a cochaperone, Hsc-70-interacting protein (Hip). BCL-2-associated athanogene-1 (BAG-1) is another cochaperone protein that can attach to Hsp70, displacing Hip from the heat shock complex [41, 42]. BAG-1 and similar polypeptides are ubiquitin-like proteins that can directly connect HSP70 and its client to the 26S proteasome. It has been postulated that BAG-1 may transport an hsp70 client close to the proteasome and guide substrates to the 26S proteasome for decomposition [43].

On the other hand, HSP47 is accumulated at many binding sites along the triple helix rather than being released at the end of procollagen folding. It could also act as an adaptor for TANGO1's procollagen loading into endoplasmic reticulum exit sites (ERESs) [44]. As a result, it is widely assumed that HSP47 is co-transported with procollagen from the ER to the ERGIC or cis-Golgi, where it is released at lower pH [45, 46].

Chaperone-mediated autophagy (CMA) and HSPs

Chaperone-mediated autophagy (CMA) is one of the primary proteolytic pathways of the lysosome-autophagy system. CMA is a type of selective autophagy in which the proteins targeted for breakdown must have a unique pentapeptide pattern recognized by HSC70/HSPA8 [47]. The chaperone-bound proteins are then delivered to lysosomes, where the lysosome-associated membrane protein type 2a (LAMP2a) receptor recognizes them [47]. CMA transports proteins for lysosomal breakdown one at a time. In contrast, autophagosomes engulf and deliver bigger structures for bulk cargo breakdown in macro-autophagy [47].

HSC and HSPs

HSCs can return to an inactive/quiescent state during liver fibrosis regression [6]. Approximately 50% of hepatic myofibroblasts escape apoptosis and revert to a quiescent-like phenotype during fibrosis recovery, downregulating fibrogenic genes and upregulating the survival proteins Hspa1a/b [6]. In fibrotic mice, transcriptional reprogramming by ectopic expression of the transcription factors FOXA3, GATA4, HNF1A, and HNF4A causes mouse myofibroblasts to transdifferentiate into hepatocyte-like cells resulting in reduced liver fibrosis [48].

ECM Remodeling and HSPs

ECM remodeling targeting is an effective method. MMP-mediated and macrophage-mediated ECM breakdown may be beneficial. Feng, Ding [49] revealed that in a mouse model of liver fibrosis, Kupfer cells (KCs) depletion delayed resolution, and adoptive transfer of KCs from WT animals expedited resolution compared to KCs from MMP9/ mice, implying that KC-derived MMP9 is required for fibrosis reversal. By interfering with collagen and elastin cross-linking, selective lysyl oxidase-like 2 (LOXL2) inhibitors diminish ECM stability and resistance to MMP destruction [50]. However, targeting LOXL2 in therapeutic studies with humanized anti-LOXL2 antibodies has so far yielded little clinical benefit [51]. Hsp47, a Col1 chaperone, was inhibited in liver fibrosis models by Hsp47 siRNA encapsulated in vitamin A-coupled liposomes, which are preferentially taken up by HSCs, showing anti-fibrotic effects [52]. COL1A1 and HSPs.

In addition to direct regulation of the COL1A1 gene, other proteins associated with collagen expressions, such as α-complex protein 2 (αCP2), transport and Golgi organization 1 (TANGO1), and HSP47, have been studied to treat liver fibrosis [53, 54]. The aberrant ECM buildup during liver fibrosis is associated with an increase in the half-life of the COL1A1 mRNA from 1.5 h in quiescent HSCs to more than 24 h in active HSCs.

Positive regulators of HSP70

Curcumin

Curcumin promotes HSP70 expression in intestinal Caco-2 cells via various signaling pathways in intestinal epithelial cells [55]. In primary rat cortical neuronal apoptosis induced by gp120 V3 loop peptide, curcumin increases HSP70 expression [56]. Hernández-Aquino, Quezada-Ramírez found that curcumin's antifibrotic actions were produced by a decrease in activated HSCs cells as a result of normalizing the GSH, NF-kB, JNK-Smad3, and TGF-β1—Smad3 pathways. Saadati, Hatami [57] found that only the curcumin group experienced significant reductions in hepatic fibrosis, serum cholesterol, glucose, and glutamic-pyruvic transaminase (ALT).

Caffeine

In Caenorhabditis elegans, coffee extract improves HS-induced HSP-70 promoter activity [58]. Using meta-analysis, Liu, Wang [59] found that consuming coffee can greatly lower your risk of developing cirrhosis and hepatic fibrosis. Modi, Feld [60] found that in all patients, including the subset with HCV infection, daily caffeine consumption above the 75(th) percentile for the cohort (308 mg) was linked to lessened liver fibrosis.

Metformin

Metformin is an antidiabetic medication used to treat and prevent the polycystic ovarian syndrome, type 2 diabetes mellitus, gestational diabetes, weight gain brought on by antipsychotics, and gestational diabetes [61]. Metformin increased the expression of numerous genes, including HSP 70 in two human esophageal squamous-cell carcinoma cell lines [62]. In Lee, Lee [63] clinical trial, a cohort of patients with metformin treatment showed a small proportion of patients developed liver fibrosis and steatosis after 2 years.

Testosterone

The primary male hormone responsible for regulating sex differentiation, producing male sex characteristics, spermatogenesis, and fertility is testosterone. In males with cirrhosis, low testosterone is a novel prognostic sign that is statistically linked to higher mortality, the requirement for transplantation, as well as risk for serious infection. [64]. The expression of HSP70-2a, HSP90, and PCNA is increased by testosterone in the experimental varicocele condition [65]. Yassin, Alwani [66] found that long-term testosterone therapy reduces hepatic steatosis and enhances liver function in hypogonadal males.

Melatonin

Melatonin is produced by the pineal gland during the night in reaction to darkness. In rats, oxidative stress is thought to contribute to functional and histopathologic changes linked to chronic cerebral hypoperfusion. Melatonin has been shown to protect against cerebral ischemia or reperfusion injury. This impact has been attributed mostly to its antioxidant characteristics which are accompanied by a rise in malondialdehyde concentration and HSP70 induction [67]. Jie, Hong [68] discovered that melatonin may reduce liver fibrosis by controlling autophagy, indicating that it may be used as a treatment for liver fibrosis. Melatonin has been shown to have antifibrotic effects on the liver, reducing profibrogenic indicators and altering some cellular functions and molecular pathways. It also acts primarily as an antioxidant and anti-inflammatory agent. [69]. Tahan, Akin [70] found that bile-duct ligation caused levels of collagen, MDA, luminal, and lucigenin to rise while GSH levels fell; however, melatonin had the opposite effect.

N-acetylcysteine (NAC)

N-acetylcysteine treatment significantly reduced hepatic inflammation and collagen deposition, decreased serum ALT, aspartate transaminase (AST), and total bilirubin, decreased hepatic hydroxyproline and malondialdehyde (MDA), down-regulated HSP47 protein expression while increasing albumin content, and significantly improved superoxide dismutase activity (SOD) [71]. Hsp70 levels increased in MG132-treated cells when NAC was added [72]. Pereira-Filho, Ferreira [73] found that through histological investigation and collagen quantification, the cirrhotic group treated with NAC demonstrated decreased degrees of fibrosis. When compared to the cirrhotic group without therapy, this group has also demonstrated less cellular membrane deterioration, less of a drop in glutathione peroxidase levels, and less expression of inducible nitric oxide synthase.

Verapamil plus bortezomib

Bortezomib strongly induced Hsp70 expression, which was enhanced when combined with verapamil in myeloma cells [74]. HSP90B1 (GRP94), HSP70, ATF6, and DDIT3 were all upregulated after verapamil and bortezomib treatment in mantle cell lymphoma [75]. In comparison to the liver fibrosis model control, verapamil caused a dose-dependent decrease in blood ALT, liver malondialdehyde, and hydroxyproline. Verapamil slowed the development of liver fibrosis and decreased hepatocyte necrosis and degeneration. Three of the verapamil-treated groups had considerably lower levels of α-SMA and TGF-β1 expression in the hepatic tissue than the liver fibrosis model control group [76]. Bortezomib is a good drug repositioning candidate since it directly decreases renal fibrosis in CKD by suppressing TGF-β1-Smad3 signaling [77].

Geranylgeranylacetone (GGA)

Geranylgeranylacetone (GGA) is an HSP70 inducer that has been used clinically as an anti-ulcer medication for many years. In the experimental traumatic brain injury mice model, GGA increased the number of HSP70+ cells [78]. In CCl4-induced liver fibrosis, GGA acted favorably by increasing the expression of HSP70. In comparison to the control group, GGA prevented liver fibrosis, reduced the amount of hydroxyproline, restored liver function, downregulated the expression of pro-fibrogenic proteins α-SMA and TGF-β1, and enhanced the expression of HSP70 [79]. Senoo, Sasaki [80] found that GGA may be used to treat liver fibrosis because it reduced fibrogenic activity, caused apoptosis in human HSCs in culture, and inhibited hepatic fibrosis in mice.

Geldanamycin analog (AAG)

HSP70 expression is increased in mouse microglia and neurons after 17-N-allylamino-17-demethoxygeldanamycin (17-AAG) treatment [81]. Zhang, Zhang [82] found that treatment with the HSP90 inhibitor 17-AAG could activate caspase-8 and caspase-9 and prevent NF-kB activation, leading to a significant increase in HSCs apoptosis. Additionally, when treated with 17-AAG, it reduced α-SMA expression and inhibited collagen synthesis induced by lipopolysaccharide and TGF-β1, suggesting that HSP90 is also involved in HSCs.

Cyclopentenone prostaglandins (cyPGs)

Cyclopentenone prostaglandins (cyPGs) have a cellular induction effect on HSP 70. HSP 70 overexpression inhibits viral infectivity factor [83]. The physiological resolution of inflammation is mediated by cyPGs, through the initiation of a genuine heat shock response (HSR), which includes cyPG-dependent activation of the HSF1 and expression of HSP70 [84].

AR-12

AR-12, derived from celecoxib, is an orally bioavailable, small-molecule inhibitor of phosphoinositide-dependent kinase-1 (PDK1) with potential antineoplastic activity. AR-12 inhibits the phosphorylation of PDK-1 and thus inhibits the activation of the serine/threonine protein kinase Akt (protein kinase B or PKB) [85], tumor cell proliferation, and inducing tumor cell death [86]. AR-12 decreased HSP90 and GRP78 protein levels while increasing HSP70 expression [87].

Zofenopril

In the chronic administration of zofenopril, an angiotensin-converting enzyme inhibitor, significant upregulation in gene expression of HSP70 was detected [88].

Anti-malaria drugs

The anti-malaria drugs quinacrine and emetine inhibited HSR in cancer cells by inducing hsp70 expression [89].

Prostaglandin E1 and lithium

Co-administration of PGE1 and lithium significantly increased cytoprotective HSP70 and HO-1 protein levels in a rat model of cerebral ischemia [90]. El-Ashmawy, Al-Ashmawy [91] found that lithium chloride promotes liver fibrosis and stimulates Wnt/β-catenin signaling. This makes the treatment for liver fibrosis using this combination less effective.

Ethanol

In the brain and liver, ethanol affects intracellular levels of GSH, HSP70, and protein carbonyls. There was a significant decrease in GSH, an increase in HSP70, and protein carbonyls in the brain, striatum, and hippocampus after seven days of ethanol treatment. Ethanol stimulates a redox mechanism that induces HSP70 induction in the brain [92].

Bleomycin

Bleomycin is an antineoplastic antibiotic that successfully induces HSP70. Bleomycin analog was reported to induce HSP70 in a pheochromocytoma cell line as well as several T-cell and monocytic cell lines. Cellular toxicity is produced by increasing the concentrations of these compounds that promote HSP70 mRNA [93]. Bleomycin produces distinct organ fibrogenesis as a net impact, even though its favorable effect on HSP70 is regarded [94].

The following figure, Fig. 1, summarizes the data regarding HSP70 positive regulators.

Fig. 1
figure 1

Positive regulators of HSP70

Negative regulators of HSP47

Aspirin

Aspirin appeared to have a protective effect against the renal damage caused by stress through its inhibitory effect on HSP60 and HSP47-mediated pathways [95, 96]. Aspirin significantly reduced liver inflammation and fibrosis through inhibition of HSC activation and proliferation, which led to a decrease in inflammatory markers such as IL-6, TNF-α, TLR4, MyD88, and NF-kB in those cells [97]. Sun, Liu [98] found that in the liver fibrosis model of rats, aspirin improved the degenerative abnormalities in the liver tissues. In a cross-sectional analysis, aspirin use was associated with significantly lower indicators of liver fibrosis in US individuals with suspected chronic liver diseases [99].

N-acetylcysteine

N-acetylcysteine had therapeutic value on liver fibrosis in the rat model [100]. Pereira-Filho, Ferreira [73] found that through histological investigation and collagen quantification, the cirrhotic group treated with NAC demonstrated decreased degree of fibrosis, less cellular membrane deterioration, and less expression of inducible nitric oxide synthase.

Pirfenidone

Pirfenidone (5-methyl-1-phenyl-2-(1H)-pyridone) is an anti-inflammatory anti-fibrotic drug that blocks the process of fibrosis in idiopathic pulmonary fibrosis patients and animal models. The antifibrotic effect of pirfenidone action is mediated through the inhibition of TGF-β1 and HSP47 expression. Xi, Li [101] found that by inhibiting Glrx, pirfenidone therapy prevents HSC activation and liver fibrosis. In HSCs, pirfenidone promotes Glrx expression in a STAT5-dependent way. Flores-Contreras, Sandoval-Rodríguez [102] found that two years of pirfenidone therapy reduces fibrosis in patients with chronic hepatitis C.

Vitamin C

Vitamin C can reduce cadmium toxicity by inhibiting changes in bioaccumulation, and hematological parameters such as calcium, magnesium, glucose, alkaline phosphatase (ALP), ALT, AST, total protein, lactate dehydrogenase (LDH), cholesterol, and lysozyme (LZM), and HSP-related genes (Hsp70, Hsp90, Hsp47, and Hsp60). Vitamin C has the potential to reduce heavy metal damage while also improving immunity [103]. Zhao and Li [104] cross-sectional study showed that there is an association of serum vitamin C with significant fibrosis in men and overweight or obese patients with NAFLD.

Tetrandrine

The in vivo comparing studies on BDL rats revealed a marked decrease in the quantification of Hsp47, collagen 1, α-SMA, and Pcol1A1 in precision-cut liver slices from fibrotic rat livers post-tetrandrine treatments [105, 106]. Hsu, Chiu [107] found that tetrandrine dramatically decreased the amount of hepatic collagen in dimethylnitrosamine-induced fibrosis in rats. Tetrandrine treatment reduced the number of NF-kB and α-SMA positive cells in the fibrotic livers. Tetrandrine therapy reduced the mRNA expression of intercellular adhesion molecule 1, α-SMA, and TGF-β1 and decreased plasma AST and ALT activity levels. Yin, Lian [108] found that tetrandrine encourages the apoptosis of activated HSCs.

Angiotensin II receptor 1 antagonist

Angiotensin II receptor 1 (AT1) antagonist candesartan maintained antifibrotic effects more effectively than ramipril in a randomized controlled prospective study involving 64 patients with chronic hepatitis C and liver fibrosis and may represent a secure and efficient therapeutic approach for liver fibrosis in patients with chronic liver diseases [109].

Dehydroepiandrosterone (DHEA)

Dehydroepiandrosterone treatment reduces the levels of androgen receptor (AR), procollagen 1 and 3, and HSP47 in the skin of postmenopausal women. DHEA significantly increased AR levels in the epidermis. A significant increase in the expression of types 1 and 3 procollagens, as well as HSP47, a procollagen chaperone protein, was observed in the dermis [110]. The potent stimulatory effect of topical DHEA on the number and size of dermal fibroblasts as well as the expression of procollagen types 1 and 3 suggests that topical DHEA may be a useful anti-aging agent in the skin [111]. Low levels of circulating DHEA-S are related to more severe NAFLD, which is denoted by the presence of NASH with the advanced fibrosis stage [112].

SB203580

SB203580 (4-(4-fluorophenyl)-2-(4-methyl sulfinyl phenyl)-5-(4-pyridyl)-imidazole) is a stress kinase inhibitor. It inhibits p38 MAPK through the block of MAPKAPK-2 activation and HSP phosphorylation [113]. Stress kinase inhibitor SB203580 downregulated collagen XVIII, CBP2/Hsp47, and VEGF expression induced by hypoxia [114]. Gao, Sun [115] found that SB203580 reduced the degree of liver fibrosis.

LY2109761

TGF-β has a significant role in metastasis and angiogenesis of cancer cells which could be inhibited by a small molecule inhibitor, LY2109761 [116]. LY2109761 has an inhibitory impact on the expression of HSP47 in rat precision-cut liver slices which is increased by the prolongation of incubation periods [117].

Imatinib

Imatinib treatment in hypertensive rats reduced PDGF-C, VEGF, HSP47, and HSP47 expression in the pulmonary veins, as well as the expression of α-SMA-positive cell proliferation [118]. When compared to traditional imatinib, HSC-targeted imatinib therapy exhibits remarkable anti-fibrotic benefits with less cytotoxicity [119]. Yoshiji, Noguchi [120] in vitro study showed that imatinib significantly reduced the proliferative and migratory effects of PDGF-BB as well as the mRNA levels of α-SMA and alpha2-(I)-procollagen in activated HSC in a dose-dependent manner. Additionally, imatinib dramatically reduced the phosphorylation of Akt, MEK1/2, and PDGFR-beta that PDGF-BB-induced in activated HSC. Unlike sorafenib, imatinib appears to merely diminish early liver fibrogenesis while not preventing long-term progression [121]. Prophylactic imatinib significantly reduced fibrosis in the first three weeks following bile duct ligation (BDL) in rats [122].

Sorafenib

Sorafenib was also found to significantly reduce the expression of fibrosis markers like α-SMA, Pcol1A1, and Hsp47 [123, 124]. Yuan, Wei [125] observed that HSC ferroptosis and ECM decrease caused by sorafenib were prevented by Fer-1 and DFO. Chen and Ji [126] observed that hepatic structure and fibrotic progression were improved and the expression of genes linked to fibrosis was dramatically decreased by sorafenib. Sorafenib prevented collagen I and α-SMA accumulation and reversed protein lysine crotonylation brought on by CCl4 injection. Pesce, Ciurleo [127] observed no additive or synergistic antifibrogenic effects for imatinib and sorafenib.

Sunitinib

Sunitinib is an indolin-2-one structural analog that is taken orally and inhibits various RTKs including VEGFR1/2/3, PDGFR, FGFR, and c-Kit [128]. Sunitinib demonstrated potent anti-tumor and anti-angiogenesis effects in a variety of cancer types in clinical trials. Sunitinib has been proven in liver fibrosis models to reduce inflammatory infiltration and the expression of fibrosis markers in liver fibrosis like HSP47 [129]. Sunitinib reduced collagen synthesis in HSCs, reduced HSC contraction, and reduced cell migration. Sunitinib inhibited the angiogenic potential of endothelial cells. Sunitinib was also found to decrease the number of VCAM-1 and ICAM-1 positive hepatic vasculature, as well as portal vein pressure, in cirrhotic rats [129]. Accordingly, Sunitinib dramatically lowers hepatic vascular density, inflammatory infiltrate, the abundance of α-SMA, LX-2 viability, collagen expression, and portal pressure in cirrhotic rats, which in turn reduces fibrosis and portal pressure as well as inflammatory infiltration.[130].

Meloxicam

Meloxicam reduced the expression of both HSP47 protein and type IV collagen mRNA which explains the improvement in mice unilateral ureteral obstruction (UUO)-induced renal interstitial fibrosis [130]. Meloxicam, a selective COX-2 inhibitor, inhibits BDL-induced hepatic fibrosis, which is accompanied by decreased hepatic TGF-β1expression and cyclooxygenase activity [131].

Emodin

Emodin is obtained mainly from Polygonaceae and is the active ingredient in Reynoutria japonica Houtt., and Rheum palmatum L. Emodin has antibacterial, antiviral, antitumor, and liver-protective properties [132, 133]. Emodin can minimize pulmonary edema and fibrosis, decrease collagen formation, and inhibit myofibroblast and inflammatory cell infiltration in the treatment of idiopathic pulmonary fibrosis (IPF). After bleomycin therapy, emodin lowered the levels of TNF-α, IL-6, TGF-β1, and HSP-47 in lung tissue [134]. Emodin can lessen the severity of liver fibrosis by decreasing the infiltration of Gr1hi monocytes and drastically reducing the production of granulin (GRN), monocyte chemoattractant protein 1 (MCP-1), TNF-α, TGF-β1, and chemokine ligand 7 (CCL7) in the liver [135].

Nintedanib and pirfenidone

Both medications influence critical regulatory levels in collagen synthesis and processing. Both drugs inhibited collagen I fibril formation and reduced and altered the appearance of collagen fibril bundles, indicating that both drugs have a completely new mechanism of action [136]. Nintedanib effectively inhibited profibrotic gene expression and collagen secretion. The regulation of the collagen chaperone FKBP10 was consistently down-regulated by nintedanib in IPF fibroblasts but not in donor fibroblasts. Pirfenidone reduced FKBP10 transcript while increasing FKBP10 protein levels in donor fibroblasts, despite not affecting FKBP10 expression in IPF fibroblasts. Nintedanib had a greater negative effect on HSP47 transcription in IPF fibroblasts than in donor fibroblasts [137, 138]. The results of the trial show that nintedanib has an antifibrotic and anti-inflammatory effect outside of the lungs. It helped lower hepatic damage, inflammation, and fibrosis in both the preventative and therapeutic treatment schedules [139].

Caveolin-1

Caveolin-1 (Cav-1) is a supporting protein that is essential for the formation of caveolae plasma membranes in most cell types. Cav-1 expression is found in most normal organs, but it is reduced when tissue is isolated or grown in culture [140]. Cav-1, due to its decreased expression in fibroblasts and monocytes, is essential in fibrosis in various tissues. Cav-1 reduced the levels of collagen I, HSP47, fibronectin, and CTGF, as well as the activation of the non-receptor tyrosine kinases Pyk2 and Src and the activation of eNOS [141].

ND-L02-s0201

NDT-05–0038 is a nuclease-resistant, synthetic, double-stranded small interfering ribonucleic acid (siRNA) developed to reversibly suppress the production of HSP47 by targeting the homologous region across humans, rats, and mice [142]. ND-L02-s0201 revealed strong antifibrotic effects and improved lung function in two robust chronic rodent models of pulmonary fibrosis, supporting its use in people with idiopathic pulmonary fibrosis [142].

Corticosteroids

Total ECM and collagen deposition were inhibited by corticosteroids via the glucocorticoid receptor and Hsp47 mRNA expression. Budesonide inhibited the mRNA expression of Hsp47 [143, 144]. Shimizu, Shimizu [145] found that an increase in corticosteroid dosage may raise the chance of developing NAFLD and liver fibrosis.

Valproic acid

Valproic acid is an anticonvulsant and mood stabilizer medication. It is widely used in the adult population to treat convulsions, migraines, and bipolar disorders. In terms of the antifibrotic investigated, sunitinib and valproic acid might lower HSP47 and PCOL1A1 gene levels [146]. AST, ALT, ALP, and GGT serum enzyme activity all increased significantly after taking valproic acid. Additionally, it markedly decreased lowered GSH content while considerably increasing MDA and NO. Valproic acid delivery simultaneously caused a significant rise in hydroxyproline, TNF-α production, and NF-kB expression, increasing the risk of liver fibrosis development [147].

The following figure, Fig. 2, summarizes the data regarding HSP47 negative regulators.

Fig. 2
figure 2

Negative regulators of HSP47

The following figure, Fig. 3, summarizes the data arranging drugs that may be most promising to treat hepatic fibrogenesis. The highest priority that approved both experimental and clinical efficacy, then that of only experimental effect and the latter types that have a conflict regarding efficacy whatever the potency against HSPs.

Fig. 3
figure 3

HSP47 and HSP70 targeted drugs. These drugs ranked according to applicability in previous studies in both experimental and clinical efficacy, then that of only experimental effect and the latter types that have a conflict regarding efficacy whatever the potency against HSPs

Conclusion

As HSP70 and HSP47 are potential targets for the control of liver fibrosis due to their role in the regulation of HSCs activation, collagen synthesis, and fibrogenesis, drugs that inhibit HSP47 or induce HSP70 can be tested for their effectiveness against liver fibrosis, especially for comorbidities.

Availability of data and materials

Not applicable.

References

  1. Berumen J, Baglieri J. Liver fibrosis: Pathophysiology and clinical implications. Mech Dis. 2021;13: e1499.

    CAS  Google Scholar 

  2. Parola M, Pinzani M. Liver fibrosis: pathophysiology, pathogenetic targets and clinical issues. Mol Aspects Med. 2019;65:37–55.

    Article  CAS  PubMed  Google Scholar 

  3. Abdelhamid AM, Youssef ME, Abd El-Fattah EE, Gobba NA, Gaafar AGA, Girgis S, et al. Blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhances the antifibrotic effect of metformin and augments its AMPK-induced NF-κB inactivation in mice intoxicated with carbon tetrachloride. Life Sci. 2021;286: 120070.

    Article  CAS  PubMed  Google Scholar 

  4. Roehlen N, Crouchet E, Baumert TF. Liver fibrosis: mechanistic concepts and therapeutic perspectives. Cells. 2020;9:875.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dhar D, Baglieri J, Kisseleva T, Brenner DA. Mechanisms of liver fibrosis and its role in liver cancer. Exp Biol Med. 2020;245:96–108.

    Article  CAS  Google Scholar 

  6. Kisseleva T, Cong M, Paik Y, Scholten D, Jiang C, Benner C, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci. 2012;109:9448–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Xie G, Diehl AM. Evidence for and against epithelial-to-mesenchymal transition in the liver. Am J Physiol Gastroint Liver Physiol. 2013;305:881–90.

    Article  Google Scholar 

  8. Koyama Y, Wang P, Liang S, Iwaisako K, Liu X, Xu J, et al. Mesothelin/mucin 16 signaling in activated portal fibroblasts regulates cholestatic liver fibrosis. J Clin Investig. 2017;127:1254–70.

    Article  PubMed  PubMed Central  Google Scholar 

  9. El-Ashmawy NE, Salem ML, Abd El-Fattah EE, Khedr EG. Targeting CD166+ lung cancer stem cells: molecular study using murine dendritic cell vaccine. Toxicol Appl Pharmacol. 2021;429: 115699.

    Article  CAS  PubMed  Google Scholar 

  10. Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: concept to treatment. J Hepatol. 2015;62:S15-24.

    Article  CAS  PubMed  Google Scholar 

  11. Aydın MM, Akçalı KC. Liver fibrosis. Turk J Gastroenterol. 2018;29:14–21.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell. 2017;66:789–800.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Caligiuri A, Bertolani C, Guerra CT, Aleffi S, Galastri S, Trappoliere M, et al. Adenosine monophosphate–activated protein kinase modulates the activated phenotype of hepatic stellate cells. Hepatology. 2008;47:668–76.

    Article  CAS  PubMed  Google Scholar 

  14. Kamada Y, Tamura S, Kiso S, Matsumoto H, Saji Y, Yoshida Y, et al. Enhanced carbon tetrachloride-induced liver fibrosis in mice lacking adiponectin. Gastroenterology. 2003;125:1796–807.

    Article  CAS  PubMed  Google Scholar 

  15. Pinzani M. Pathophysiology of liver fibrosis. Digestive Dis. 2015;33:492–7.

    Article  Google Scholar 

  16. Abd El-Fattah EE, Abdelhamid AM. Benzo[a]pyrene immunogenetics and immune archetype reprogramming of lung. Toxicology. 2021;463: 152994.

    Article  CAS  PubMed  Google Scholar 

  17. Abdelhamid AM, Saber S, Youssef ME, Gaafar AGA, Eissa H, Abd-Eldayem MA, et al. Empagliflozin adjunct with metformin for the inhibition of hepatocellular carcinoma progression: Emerging approach for new application. Biomed Pharmacother. 2022;145: 112455.

    Article  CAS  PubMed  Google Scholar 

  18. Tan Z, Sun H, Xue T, Gan C, Liu H, Xie Y, et al. Liver fibrosis: therapeutic targets and advances in drug therapy. Front Cell Develo Biol. 2021;9: 730176.

    Article  Google Scholar 

  19. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18:151–66.

    Article  PubMed  Google Scholar 

  20. Tutar L, Tutar Y. Heat shock proteins; an overview. Curr Pharm Biotechnol. 2010;11:216–22.

    Article  CAS  PubMed  Google Scholar 

  21. Zininga T, Ramatsui L, Shonhai A. Heat shock proteins as immunomodulants. Molecules. 2018;23(11):2846.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Tanguy J, Pommerolle L, Garrido C, Kolb M, Bonniaud P, Goirand F, et al. Extracellular heat shock proteins as therapeutic targets and biomarkers in fibrosing interstitial lung diseases. Intern J Mol Sci. 2021;22:9316.

    Article  CAS  Google Scholar 

  23. Esmaeili Z, Niaz Q, Saffari PM, Dehpour AR, Rezayat SM, Jazaeri F. Evaluation of the effect of heat shock protein 70 targeted drugs on cirrhotic cardiomyopathy in biliary cirrhotic rats. Life Sci. 2021;273: 119261.

    Article  CAS  PubMed  Google Scholar 

  24. Gehrmann M, Cervello M, Montalto G, Cappello F, Gulino A, Knape C, et al. Heat shock protein 70 serum levels differ significantly in patients with chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Front Immunol. 2014;5:307.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Sellares J, Veraldi KL. Intracellular heat shock protein 70 deficiency in pulmonary fibrosis. Am J Respir Cell Mol Biol. 2019;60:629–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhao Y, Dang Z, Xu S, Chong S. Heat shock protein 47 effects on hepatic stellate cell-associated receptors in hepatic fibrosis of Schistosoma japonicum-infected mice. Biol Chem. 2017;398:1357–66.

    Article  CAS  PubMed  Google Scholar 

  27. Huang JQ, Tao R, Li L, Ma K, Xu L, Ai G, et al. Involvement of heat shock protein 47 in Schistosoma japonicum-induced hepatic fibrosis in mice. Int J Parasitol. 2014;44:23–35.

    Article  CAS  PubMed  Google Scholar 

  28. Salem ML, El-Ashmawy NE, Abd El-Fattah EE, Khedr EG. Immunosuppressive role of Benzo[a]pyrene in induction of lung cancer in mice. Chem Biol Interact. 2021;333: 109330.

    Article  CAS  PubMed  Google Scholar 

  29. Lønsmann I, Gudmann NS, Manon-Jensen T, Thiele M, Moreno YM, Langholm LL, et al. Serologically assessed heat shock protein 47 is related to fibrosis stage in early compensated alcohol-related liver disease. Clin Biochem. 2022;104:36–43.

    Article  PubMed  Google Scholar 

  30. Li L, Wu T, Huang J, Ma K, Xu L, Wang H, et al. Expression of heat shock protein 47, transforming growth factor-beta 1, and connective tissue growth factor in liver tissue of patients with Schistosoma japonicum-induced hepatic fibrosis. Parasitology. 2015;142:341–51.

    Article  CAS  PubMed  Google Scholar 

  31. Sato Y, Yoneda A, Shimizu F, Nishimura M, Shimoyama R, Tashiro Y, et al. Resolution of fibrosis by siRNA HSP47 in vitamin a-coupled liposomes induces regeneration of chronically injured livers. J Gastroenterol Hepatol. 2021;36:3418–28.

    Article  CAS  PubMed  Google Scholar 

  32. Huang Y, Lu J, Xu Y, Xiong C, Tong D, Hu N, et al. Xiaochaihu decorction relieves liver fibrosis caused by Schistosoma japonicum infection via the HSP47/TGF-β pathway. Parasit Vectors. 2020;13:254.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Kurumi H, Takata T, Kanda T, Sugihara T, Kakugawa T, Yokota SI, et al. Investigating the role of heat shock protein 47 in fibrosis in Crohn’s disease. Sci Rep. 2022;12:10966.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rizk FH, Sarhan NI, Soliman NA, Ibrahim MAA, Abd-Elsalam M, Abd-Elsalam S. Heat shock protein 47 as indispensible participant in liver fibrosis: Possible protective effect of lactoferrin. IUBMB Life. 2018;70:795–805.

    Article  CAS  PubMed  Google Scholar 

  35. Wei S, Wang Q, Zhou H, Qiu J, Li C, Shi C, et al. miR-455-3p alleviates hepatic stellate cell activation and liver fibrosis by suppressing HSF1 expression. Molecular Therapy Nucleic Acids. 2019;16:758–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li L, Zhang L, Zhao X, Cao J, Li J, Chu G. Downregulation of miR-152 contributes to the progression of liver fibrosis via targeting Gli3 in vivo and in vitro. Exp Ther Med. 2019;18:425–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang Y, Ye W-L, Zhang R-N, He X-S, Wang J-R, Liu Y-X, et al. The role of TGF-β signaling pathways in cancer and its potential as a therapeutic target. Evidence Based Comply and Altern Med. 2021. https://doi.org/10.1155/2021/6675208.

    Article  Google Scholar 

  38. Zhang K, Han X, Zhang Z, Zheng L, Hu Z, Yao Q, et al. The liver-enriched lnc-LFAR1 promotes liver fibrosis by activating TGFβ and Notch pathways. Nat Commun. 2017;8:1–16.

    Google Scholar 

  39. Lin X, Duan X, Liang Y-Y, Su Y, Wrighton KH, Long J, et al. PPM1A functions as a Smad phosphatase to terminate TGFβ signaling. Cell. 2006;125:915–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Sever R, Glass CK. Signaling by nuclear receptors. Cold Spring Harb Perspect Biol. 2013;5: a016709.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Hohfeld J, Jentsch S. GrpE-like regulation of the Hsc70 chaperone by the anti-apoptotic protein BAG-1. Embo J. 1998;17:847.

    Article  Google Scholar 

  42. Kanelakis KC, Morishima Y, Dittmar KD, Galigniana MD, Takayama S, Reed JC, et al. Differential effects of the hsp70-binding protein BAG-1 on glucocorticoid receptor folding by the hsp90-based chaperone machinery. J Biol Chem. 1999;274:34134–40.

    Article  CAS  PubMed  Google Scholar 

  43. Alarid ET. Lives and times of nuclear receptors. Mol Endocrinol. 2006;20:1972–81.

    Article  CAS  PubMed  Google Scholar 

  44. Ishikawa Y, Ito S, Nagata K, Sakai LY, Bächinger HP. Intracellular mechanisms of molecular recognition and sorting for transport of large extracellular matrix molecules. Proc Natl Acad Sci. 2016;113:E6036–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Oecal S, Socher E, Uthoff M, Ernst C, Zaucke F, Sticht H, et al. The pH-dependent client release from the collagen-specific chaperone HSP47 is triggered by a tandem histidine pair. J Biol Chem. 2016;291:12612–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Saga S, Nagata K, Chen W-T, Yamada KM. pH-dependent function, purification, and intracellular location of a major collagen-binding glycoprotein. J Cell Biol. 1987;105:517–27.

    Article  CAS  PubMed  Google Scholar 

  47. Zhang M, Serna-Salas S, Damba T, Borghesan M, Demaria M, Moshage H. Hepatic stellate cell senescence in liver fibrosis: Characteristics, mechanisms and perspectives. Mech Ageing Dev. 2021;199: 111572.

    Article  CAS  PubMed  Google Scholar 

  48. Song G, Pacher M, Balakrishnan A, Yuan Q, Tsay H-C, Yang D, et al. Direct reprogramming of hepatic myofibroblasts into hepatocytes in vivo attenuates liver fibrosis. Cell Stem Cell. 2016;18:797–808.

    Article  CAS  PubMed  Google Scholar 

  49. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ikenaga N, Peng Z-W, Vaid KA, Liu SB, Yoshida S, Sverdlov DY, et al. Selective targeting of lysyl oxidase-like 2 (LOXL2) suppresses hepatic fibrosis progression and accelerates its reversal. Gut. 2017;66:1697–708.

    Article  CAS  PubMed  Google Scholar 

  51. Harrison SA, Abdelmalek MF, Caldwell S, Shiffman ML, Diehl AM, Ghalib R, et al. Simtuzumab is ineffective for patients with bridging fibrosis or compensated cirrhosis caused by nonalcoholic steatohepatitis. Gastroenterology. 2018;155:1140–53.

    Article  CAS  PubMed  Google Scholar 

  52. Sato Y, Murase K, Kato J, Kobune M, Sato T, Kawano Y, et al. Resolution of liver cirrhosis using vitamin A–coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol. 2008;26:431–42.

    Article  CAS  PubMed  Google Scholar 

  53. Liu H, Chen Z, Jin W, Barve A, Wan Y-JY, Cheng K. Silencing of α-complex protein-2 reverses alcohol-and cytokine-induced fibrogenesis in hepatic stellate cells. Liver Res. 2017;1:70–9.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Maiers JL, Kostallari E, Mushref M, deAssuncao TM, Li H, Jalan-Sakrikar N, et al. The unfolded protein response mediates fibrogenesis and collagen I secretion through regulating TANGO1 in mice. Hepatology. 2017;65:983–98.

    Article  CAS  PubMed  Google Scholar 

  55. Guo M, Xu W, Yamamoto Y, Suzuki T. Curcumin increases heat shock protein 70 expression via different signaling pathways in intestinal epithelial cells. Arch Biochem Biophys. 2021;707: 108938.

    Article  CAS  PubMed  Google Scholar 

  56. Xia C, Cai Y, Li S, Yang J, Xiao G. Curcumin increases HSP70 expression in primary rat cortical neuronal apoptosis induced by gp120 V3 Loop Peptide. Neurochem Res. 2015;40:1996–2005.

    Article  CAS  PubMed  Google Scholar 

  57. Saadati S, Hatami B, Yari Z, Shahrbaf MA, Eghtesad S, Mansour A, et al. The effects of curcumin supplementation on liver enzymes, lipid profile, glucose homeostasis, and hepatic steatosis and fibrosis in patients with non-alcoholic fatty liver disease. Eur J Clin Nutr. 2019;73:441–9.

    Article  CAS  PubMed  Google Scholar 

  58. Brunquell J, Morris S, Snyder A, Westerheide SD. Coffee extract and caffeine enhance the heat shock response and promote proteostasis in an HSF-1-dependent manner in Caenorhabditis elegans. Cell Stress Chaperones. 2018;23:65–75.

    Article  CAS  PubMed  Google Scholar 

  59. Liu F, Wang X, Wu G, Chen L, Hu P, Ren H, et al. Coffee consumption decreases risks for hepatic fibrosis and cirrhosis: a meta-analysis. PLoS ONE. 2015;10: e0142457.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Modi AA, Feld JJ, Park Y, Kleiner DE, Everhart JE, Liang TJ, et al. Increased caffeine consumption is associated with reduced hepatic fibrosis. Hepatology. 2010;51:201–9.

    Article  CAS  PubMed  Google Scholar 

  61. Abd El-Fattah EE, Zakaria AY. Metformin modulate immune fitness in hepatocellular carcinoma: Molecular and cellular approach. Int Immunopharmacol. 2022;109: 108889.

    Article  CAS  PubMed  Google Scholar 

  62. Sekino N, Kano M, Kobayashi S, Murakami K, Sakata H, Toyozumi T, et al. Metformin-induced heat shock protein family a member 6 is a promising biomarker of esophageal squamous cell carcinoma. Oncology. 2022;100:267–77.

    Article  CAS  PubMed  Google Scholar 

  63. Lee HW, Lee JS, Kim BK, Park JY, Kim DY, Ahn SH, et al. Evolution of liver fibrosis and steatosis markers in patients with type 2 diabetes after metformin treatment for 2 years. J Diabetes Complications. 2021;35: 107747.

    Article  CAS  PubMed  Google Scholar 

  64. Copyright © 2022, StatPearls Publishing LLC. 2022.

  65. Sinclair M, Gow PJ, Grossmann M, Shannon A, Hoermann R, Angus PW. Low serum testosterone is associated with adverse outcome in men with cirrhosis independent of the model for end-stage liver disease score. Liver Transpl. 2016;22:1482–90.

    Article  PubMed  Google Scholar 

  66. Salmani S, Razi M, Sarrafzadeh-Rezaei F, Mahmoudian A. Testosterone amplifies HSP70-2a, HSP90 and PCNA expression in experimental varicocele condition: Implication for DNA fragmentation. Reprod Biol. 2020;20:384–95.

    Article  PubMed  Google Scholar 

  67. Yassin AA, Alwani M, Talib R, Almehmadi Y, Nettleship JE, Alrumaihi K, et al. Long-term testosterone therapy improves liver parameters and steatosis in hypogonadal men: a prospective controlled registry study. Aging Male. 2020;23:1553–63.

    Article  PubMed  Google Scholar 

  68. Ozacmak VH, Barut F, Ozacmak HS. Melatonin provides neuroprotection by reducing oxidative stress and HSP70 expression during chronic cerebral hypoperfusion in ovariectomized rats. J Pineal Res. 2009;47:156–63.

    Article  CAS  PubMed  Google Scholar 

  69. Jie L, Hong R-t, Zhang Y-j, Sha L-l, Chen W, Ren X-f. Melatonin alleviates liver fibrosis by inhibiting autophagy. Current Med Sci. 2022;42:498–504.

    Article  CAS  Google Scholar 

  70. San-Miguel B, Fernández-Palanca P, Mauriz JL, Tuñón MJ, González-Gallego J. Beneficial effects of melatonin on liver fibrosis: a systematic review of current biological evidence. J Cell Physiol. 2022;237:2740–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Tahan G, Akin H, Aydogan F, Ramadan SS, Yapicier O, Tarcin O, et al. Melatonin ameliorates liver fibrosis induced by bile-duct ligation in rats. Canad J Surg. 2010;53:313–8.

    PubMed  PubMed Central  Google Scholar 

  72. Abd El-Fattah EE. IDO/kynurenine pathway in cancer: possible therapeutic approaches. J Transl Med. 2022;20:347.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Jiang Y, Rumble JL, Gleixner AM, Unnithan AS, Pulugulla SH, Posimo JM, et al. N-Acetyl cysteine blunts proteotoxicity in a heat shock protein-dependent manner. Neuroscience. 2013;255:19–32.

    Article  CAS  PubMed  Google Scholar 

  74. Pereira-Filho G, Ferreira C, Schwengber A, Marroni C, Zettler C, Marroni N. Role of N-acetylcysteine on fibrosis and oxidative stress in cirrhotic rats. Arq Gastroenterol. 2008;45:156–62.

    Article  PubMed  Google Scholar 

  75. Meister S, Frey B, Lang VR, Gaipl US, Schett G, Schlötzer-Schrehardt U, et al. Calcium channel blocker verapamil enhances endoplasmic reticulum stress and cell death induced by proteasome inhibition in myeloma cells. Neoplasia. 2010;12:550–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Chen Z, Romaguera J, Wang M, Fayad L, Kwak LW, McCarty N. Verapamil synergistically enhances cytotoxicity of bortezomib in mantle cell lymphoma via induction of reactive oxygen species production. Br J Haematol. 2012;159:243–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Xu D, Wu Y, Liao Z-X, Wang H. Protective effect of verapamil on multiple hepatotoxic factors-induced liver fibrosis in rats. Pharmacol Res. 2007;55:280–6.

    Article  CAS  PubMed  Google Scholar 

  78. Zeniya M, Mori T, Yui N, Nomura N, Mandai S, Isobe K, et al. The proteasome inhibitor bortezomib attenuates renal fibrosis in mice via the suppression of TGF-β1. Sci Rep. 2017;7:13086.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Zhao Z, Faden AI, Loane DJ, Lipinski MM, Sabirzhanov B, Stoica BA. Neuroprotective effects of geranylgeranylacetone in experimental traumatic brain injury. J Cerebral Blood Flow Metabol. 2013;33:1897–908.

    Article  CAS  Google Scholar 

  80. He W, Zhuang Y, Wang L, Qi L, Chen B, Wang M, et al. Geranylgeranylacetone attenuates hepatic fibrosis by increasing the expression of heat shock protein 70. Mol Med Rep. 2015;12:4895–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Senoo T, Sasaki R, Akazawa Y, Ichikawa T, Miuma S, Miyaaki H, et al. Geranylgeranylacetone attenuates fibrogenic activity and induces apoptosis in cultured human hepatic stellate cells and reduces liver fibrosis in carbon tetrachloride-treated mice. BMC Gastroenterol. 2018;18:34.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Kim N, Kim JY, Yenari MA. Pharmacological induction of the 70-kDa heat shock protein protects against brain injury. Neuroscience. 2015;284:912–9.

    Article  CAS  PubMed  Google Scholar 

  83. Zhang X, Zhang X, Huang W, Ge X. The role of heat shock proteins in the regulation of fibrotic diseases. Biomed Pharmacother. 2021;135: 111067.

    Article  CAS  PubMed  Google Scholar 

  84. Sugiyama R, Abe M, Nishitsuji H, Murakami Y, Takeuchi H, Takaku H. Induction of heat-shock protein 70 by prostaglandin A1 inhibits HIV-1 Vif-mediated degradation of APOBEC3G. Antiviral Res. 2013;99:307–11.

    Article  CAS  PubMed  Google Scholar 

  85. Gutierrez LLP, Marques CV, Scomazzon SP, Schroeder HT, Fernandes JR, da Silva RJ, et al. A-family anti-inflammatory cyclopentenone prostaglandins: a novel class of non-statin inhibitors of HMG-CoA reductase. Biochimie. 2021;182:37–50.

    Article  CAS  PubMed  Google Scholar 

  86. Abd El-Fattah EE, Saber S, Youssef ME, Eissa H, El-Ahwany E, Amin NA, et al. AKT-AMPKα-mTOR-dependent HIF-1α activation is a new therapeutic target for cancer treatment: a novel approach to repositioning the antidiabetic drug sitagliptin for the management of hepatocellular carcinoma. Front Pharmacol. 2021;12: 720173.

    Article  CAS  PubMed  Google Scholar 

  87. Abdulrahman BA, Abdelaziz D, Thapa S, Lu L, Jain S, Gilch S, et al. The celecoxib derivatives AR-12 and AR-14 induce autophagy and clear prion-infected cells from prions. Sci Rep. 2017;7:17565.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Booth L, Shuch B, Albers T, Roberts JL, Tavallai M, Proniuk S, et al. Multi-kinase inhibitors can associate with heat shock proteins through their NH2-termini by which they suppress chaperone function. Oncotarget. 2016;7:12975–96.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Carnicelli V, Frascarelli S, Zucchi R. Effect of acute and chronic zofenopril administration on cardiac gene expression. Mol Cell Biochem. 2011;352:301–7.

    Article  CAS  PubMed  Google Scholar 

  90. Neznanov N, Gorbachev AV, Neznanova L, Komarov AP, Gurova KV, Gasparian AV, et al. Anti-malaria drug blocks proteotoxic stress response: anti-cancer implications. Cell Cycle. 2009;8:3960–70.

    Article  CAS  PubMed  Google Scholar 

  91. Han R, Gao B, Sheng R, Zhang LS, Zhang HL, Gu ZL, et al. Synergistic effects of prostaglandin E1 and lithium in a rat model of cerebral ischemia. Acta Pharmacol Sin. 2008;29:1141–9.

    Article  CAS  PubMed  Google Scholar 

  92. El-Ashmawy NE, Al-Ashmawy GM, Fakher HE, Khedr NF. The role of WNT/β-catenin signaling pathway and glutamine metabolism in the pathogenesis of CCl4-induced liver fibrosis: Repositioning of niclosamide and concerns about lithium. Cytokine. 2020;136: 155250.

    Article  CAS  PubMed  Google Scholar 

  93. Liu J. Ethanol and liver: recent insights into the mechanisms of ethanol-induced fatty liver. World J Gastroenterol. 2014;20:14672–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Ali TFS, Taira N, Iwamaru K, Koga R, Kamo M, Radwan MO, et al. HSP70 induction by bleomycin metal core analogs. Bioorg Med Chem Lett. 2020;30: 127002.

    Article  CAS  PubMed  Google Scholar 

  95. Vásquez-Garzón VR, Ramírez-Cosmes A, Reyes-Jiménez E, Carrasco-Torres G, Hernández-García S, Aguilar-Ruiz SR, et al. Liver damage in bleomycin-induced pulmonary fibrosis in mice. Naunyn Schmiedebergs Arch Pharmacol. 2019;392:1503–13.

    Article  PubMed  Google Scholar 

  96. Wu D, Zhang M, Xu J, Song E, Lv Y, Tang S, et al. In vitro evaluation of aspirin-induced HspB1 against heat stress damage in chicken myocardial cells. Cell Stress Chaperones. 2016;21:405–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Tang S, Zhou S, Yin B, Xu J, Di L, Zhang J, et al. Heat stress-induced renal damage in poultry and the protective effects of HSP60 and HSP47. Cell Stress Chaperones. 2018;23:1033–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Liu Y, Nong L, Jia Y, Tan A, Duan L, Lu Y, et al. Aspirin alleviates hepatic fibrosis by suppressing hepatic stellate cells activation via the TLR4/NF-κB pathway. Aging. 2020;12:6058–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Sun Y, Liu B, Xie J, Jiang X, Xiao B, Hu X, et al. Aspirin attenuates liver fibrosis by suppressing TGF-β1/Smad signaling. Mol Med Rep. 2022;25:181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Jiang ZG, Feldbrügge L, Tapper EB, Popov Y, Ghaziani T, Afdhal N, et al. Aspirin use is associated with lower indices of liver fibrosis among adults in the United States. Aliment Pharmacol Ther. 2016;43:734–43.

    Article  CAS  PubMed  Google Scholar 

  101. Nakayama S, Mukae H, Sakamoto N, Kakugawa T, Yoshioka S, Soda H, et al. Pirfenidone inhibits the expression of HSP47 in TGF-β1-stimulated human lung fibroblasts. Life Sci. 2008;82:210–7.

    Article  CAS  PubMed  Google Scholar 

  102. Xi Y, Li Y, Xu P, Li S, Liu Z, Tung H-c, et al. The anti-fibrotic drug pirfenidone inhibits liver fibrosis by targeting the small oxidoreductase glutaredoxin-1. Sci Adv. 2021;7:eabg9241.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Flores-Contreras L, Sandoval-Rodríguez AS, Mena-Enriquez MG, Lucano-Landeros S, Arellano-Olivera I, Álvarez-Álvarez A, et al. Treatment with pirfenidone for two years decreases fibrosis, cytokine levels and enhances CB2 gene expression in patients with chronic hepatitis C. BMC Gastroenterol. 2014;14:131.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Wang H, Feng Y, Ming M, Song J, Chen Z, Xiao Z. Amelioration of Cd-induced bioaccumulation, hematological parameters, and heat shock protein-related genes by Vitamin C on common carp. Compar Biochem Physiol Toxicol Pharmacol. 2022;1:109362.

    Article  Google Scholar 

  105. Zhao Y, Li H. Association of serum vitamin C with liver fibrosis in adults with nonalcoholic fatty liver disease. Scand J Gastroenterol. 2022;57:872–7.

    Article  CAS  PubMed  Google Scholar 

  106. Hsu Y-C, Chiu Y-T, Lee C-Y, Wu C-F, Huang Y-T. Anti-fibrotic effects of tetrandrine on bile-duct ligated rats. Can J Physiol Pharmacol. 2006;84:967–76.

    Article  CAS  PubMed  Google Scholar 

  107. Park PH, Nan JX, Park EJ, Kang HC, Kim JY, Ko G, et al. Effect of tetrandrine on experimental hepatic fibrosis induced by bile duct ligation and scission in rats. Pharmacol Toxicol. 2000;87:261–8.

    Article  CAS  PubMed  Google Scholar 

  108. Hsu Y-C, Chiu Y-T, Cheng C-C, Wu C-F, Lin Y-L, Huang Y-T. Antifibrotic effects of tetrandrine on hepatic stellate cells and rats with liver fibrosis. J Gastroenterol Hepatol. 2007;22:99–111.

    Article  CAS  PubMed  Google Scholar 

  109. Yin MF, Lian LH, Piao DM, Nan JX. Tetrandrine stimulates the apoptosis of hepatic stellate cells and ameliorates development of fibrosis in a thioacetamide rat model. World J Gastroenterol. 2007;13:1214–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Mostafa TM, El-azab GA, Badra GA, Abdelwahed AS, Elsayed AA. Effect of candesartan and ramipril on liver fibrosis in patients with chronic hepatitis C viral infection: a randomized controlled prospective study. Curr Ther Res. 2021;95: 100654.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Dafforn TR, Della M, Miller AD. The molecular interactions of heat shock protein 47 (Hsp47) and their implications for collagen biosynthesis. J Biol Chem. 2001;276:49310–9.

    Article  CAS  PubMed  Google Scholar 

  112. El-Alfy M, Deloche C, Azzi L, Bernard B, Bernerd F, Coutet J, et al. Skin responses to topical dehydroepiandrosterone: implications in antiageing treatment? Br J Dermatol. 2010;163:968–76.

    Article  CAS  PubMed  Google Scholar 

  113. Charlton M, Angulo P, Chalasani N, Merriman R, Viker K, Charatcharoenwitthaya P, et al. Low circulating levels of dehydroepiandrosterone in histologically advanced nonalcoholic fatty liver disease. Hepatology. 2008;47:484–92.

    Article  CAS  PubMed  Google Scholar 

  114. Cuenda A, Rouse J, Doza YN, Meier R, Cohen P, Gallagher TF, et al. SB 203580 is a specific inhibitor of a MAP kinase homologue which is stimulated by cellular stresses and interleukin-1. FEBS Lett. 1995;364:229–33.

    Article  CAS  PubMed  Google Scholar 

  115. Stewart J, Siavash H, Hebert C, Norris K, Nikitakis N, Sauk J. Phenotypic switching of VEGF and collagen XVIII during hypoxia in head and neck squamous carcinoma cells. Oral Oncol. 2003;39:862–9.

    Article  CAS  PubMed  Google Scholar 

  116. Gao B, Sun W, Meng X, Xue D, Zhang W. Screening of differentially expressed protein kinases in bone marrow endothelial cells and the protective effects of the p38a inhibitor SB203580 on bone marrow in liver fibrosis. Mol Med Rep. 2016;14:4629–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Melisi D, Ishiyama S, Sclabas GM, Fleming JB, Xia Q, Tortora G, et al. LY2109761, a novel transforming growth factor β receptor type I and type II dual inhibitor, as a therapeutic approach to suppressing pancreatic cancer metastasis. Mol Cancer Ther. 2008;7:829–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Ishida Y, Nagata K. Hsp47 as a collagen-specific molecular chaperone Methods in enzymology. Amsterdam: Elsevier; 2011.

    Google Scholar 

  119. Iwasaki Y-k, Yamashita T, Sekiguchi A, Hayami N, Shimizu W. Importance of pulmonary vein preferential fibrosis for atrial fibrillation promotion in hypertensive rat hearts. Canad J Cardiol. 2016;32:767–76.

    Article  Google Scholar 

  120. El-Mezayen NS, El-Hadidy WF, El-Refaie WM, Shalaby TI, Khattab MM, El-Khatib AS. Hepatic stellate cell-targeted imatinib nanomedicine versus conventional imatinib: a novel strategy with potent efficacy in experimental liver fibrosis. J Control Release. 2017;266:226–37.

    Article  CAS  PubMed  Google Scholar 

  121. Yoshiji H, Noguchi R, Kuriyama S, Ikenaka Y, Yoshii J, Yanase K, et al. Imatinib mesylate (STI-571) attenuates liver fibrosis development in rats. Am J Physiol Gastrointest Liver Physiol. 2005;288:G907–13.

    Article  CAS  PubMed  Google Scholar 

  122. Neef M, Ledermann M, Saegesser H, Schneider V, Widmer N, Decosterd LA, et al. Oral imatinib treatment reduces early fibrogenesis but does not prevent progression in the long term. J Hepatol. 2006;44:167–75.

    Article  CAS  PubMed  Google Scholar 

  123. Qu K, Huang Z, Lin T, Liu S, Chang H, Yan Z, et al. New insight into the anti-liver fibrosis effect of multitargeted tyrosine kinase inhibitors: from molecular target to clinical trials. Front Pharmacol. 2016;6:300.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Westra IM, Oosterhuis D, Groothuis GM, Olinga P. The effect of antifibrotic drugs in rat precision-cut fibrotic liver slices. PLoS ONE. 2014;9: e95462.

    Article  PubMed  PubMed Central  Google Scholar 

  125. El-Ashmawy NE, Khedr EG, El-Bahrawy HA, Abd El-Fattah EE. Sorafenib effect on liver neoplastic changes in rats: more than a kinase inhibitor. Clin Exp Med. 2017;17:185–91.

    Article  CAS  PubMed  Google Scholar 

  126. Yuan S, Wei C, Liu G, Zhang L, Li J, Li L, et al. Sorafenib attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis via HIF-1α/SLC7A11 pathway. Cell Prolif. 2022;55: e13158.

    Article  CAS  PubMed  Google Scholar 

  127. Chen XF, Ji S. Sorafenib attenuates fibrotic hepatic injury through mediating lysine crotonylation. Drug Des Dev Ther. 2022;16:2133–44.

    Article  Google Scholar 

  128. Pesce A, Ciurleo R, Bramanti A, Armeli Iapichino EC, Petralia MC, Magro GG, et al. Effects of combined admistration of imatinib and sorafenib in a murine model of liver fibrosis. Molecules. 2020;25(18):4310.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Faivre S, Demetri G, Sargent W, Raymond E. Molecular basis for sunitinib efficacy and future clinical development. Nat Rev Drug Discovery. 2007;6:734–45.

    Article  CAS  PubMed  Google Scholar 

  130. Tugues S, Fernandez-Varo G, Muñoz-Luque J, Ros J, Arroyo V, Rodés J, et al. Antiangiogenic treatment with sunitinib ameliorates inflammatory infiltrate, fibrosis, and portal pressure in cirrhotic rats. Hepatology. 2007;46:1919–26.

    Article  CAS  PubMed  Google Scholar 

  131. Honma S, Shinohara M, Takahashi N, Nakamura K, Hamano S, Mitazaki S, et al. Effect of cyclooxygenase (COX)-2 inhibition on mouse renal interstitial fibrosis. Eur J Pharmacol. 2014;740:578–83.

    Article  CAS  PubMed  Google Scholar 

  132. Kim SM, Park KC, Kim HG, Han SJ. Effect of selective cyclooxygenase-2 inhibitor meloxicam on liver fibrosis in rats with ligated common bile ducts. Hepatol Res. 2008;38:800–9.

    Article  CAS  PubMed  Google Scholar 

  133. Li L, Song X, Yin Z, Jia R, Li Z, Zhou X, et al. The antibacterial activity and action mechanism of emodin from Polygonum cuspidatum against Haemophilus parasuis in vitro. Microbiol Res. 2016;186:139–45.

    Article  PubMed  Google Scholar 

  134. Lin W, Wang C, Ling C. Research progress in anti-tumor effect of emodin. Zhongguo Zhong yao za zhi= Zhongguo zhongyao zazhi. China J Chin Materia Med. 2015;40:3937–40.

    CAS  Google Scholar 

  135. Guan R, Zhao X, Wang X, Song N, Guo Y, Yan X, et al. Emodin alleviates bleomycin-induced pulmonary fibrosis in rats. Toxicol Lett. 2016;262:161–72.

    Article  CAS  PubMed  Google Scholar 

  136. Zhao XA, Chen G, Liu Y, Wu H, Chen J, Xiong Y, et al. Emodin alleviates liver fibrosis of mice by reducing infiltration of gr1(hi) monocytes. Evidence Based Complem Alter Med. 2018;2018:5738101.

    Article  Google Scholar 

  137. Knüppel L, Ishikawa Y, Aichler M, Heinzelmann K, Hatz R, Behr J, et al. A novel antifibrotic mechanism of nintedanib and pirfenidone Inhibition of collagen fibril assembly. Am J Respir Cell Mol Biol. 2017;57:77–90.

    Article  PubMed  Google Scholar 

  138. Hostettler KE, Zhong J, Papakonstantinou E, Karakiulakis G, Tamm M, Seidel P, et al. Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis. Respir Res. 2014;15:1–9.

    Article  Google Scholar 

  139. Staab-Weijnitz CA, Fernandez IE, Knüppel L, Maul J, Heinzelmann K, Juan-Guardela BM, et al. FK506-binding protein 10, a potential novel drug target for idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2015;192:455–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Wollin L, Togbe D, Ryffel B. Effects of nintedanib in an animal model of liver fibrosis. Biomed Res Int. 2020;2020:3867198.

    Article  PubMed  PubMed Central  Google Scholar 

  141. Carver LA, Schnitzer JE. Caveolae: mining little caves for new cancer targets. Nat Rev Cancer. 2003;3:571–81.

    Article  CAS  PubMed  Google Scholar 

  142. Pleasant-Jenkins D, Reese C, Chinnakkannu P, Kasiganesan H, Tourkina E, Hoffman S, et al. Reversal of maladaptive fibrosis and compromised ventricular function in the pressure overloaded heart by a caveolin-1 surrogate peptide. Lab Invest. 2017;97:370–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Liu Y, Liu J, Quimbo A, Xia F, Yao J, Clamme J-P, et al. Anti-HSP47 siRNA lipid nanoparticle ND-L02-s0201 reverses interstitial pulmonary fibrosis in preclinical rat models. ERJ open research 2021;7.

  144. Kuroda K, Tsukifuji R, Shinkai H. Increased expression of heat-shock protein 47 is associated with overproduction of type I procollagen in systemic sclerosis skin fibroblasts. J Investig Dermatol. 1998;111:1023–8.

    Article  CAS  PubMed  Google Scholar 

  145. Kakugawa T, Yokota S-i, Ishimatsu Y, Hayashi T, Nakashima S, Hara S, et al. Serum heat shock protein 47 levels are elevated in acute exacerbation of idiopathic pulmonary fibrosis. Cell Stress Chaper. 2013;18:581–90.

    Article  CAS  Google Scholar 

  146. Shimizu H, Shimizu T, Takahashi D, Asano T, Arai R, Takakuwa Y, et al. Corticosteroid dose increase is a risk factor for nonalcoholic fatty liver disease and contralateral osteonecrosis of the femoral head: a case report. BMC Musculoskelet Disord. 2019;20:88.

    Article  PubMed  PubMed Central  Google Scholar 

  147. Westra IM, Mutsaers HA, Luangmonkong T, Hadi M, Oosterhuis D, de Jong KP, et al. Human precision-cut liver slices as a model to test antifibrotic drugs in the early onset of liver fibrosis. Toxicol In Vitro. 2016;35:77–85.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).

Author information

Authors and Affiliations

Authors

Contributions

EEAEF Conceptualization, Methodology, Reviewing, Supervising and Editing. AYZ Investigation and Writing- Original draft preparation. Both authors read and approved the final manuscript.

Corresponding author

Correspondence to Eslam E. Abd El-Fattah.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that there is no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Abd El-Fattah, E.E., Zakaria, A.Y. Targeting HSP47 and HSP70: promising therapeutic approaches in liver fibrosis management. J Transl Med 20, 544 (2022). https://doi.org/10.1186/s12967-022-03759-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-022-03759-z

Keywords