ArticlePublisher preview availableLiterature Review

Regulation of wound healing and fibrosis by galectins

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Galectins are a family of proteins with at least one carbohydrate-recognition domain. Galectins are present in various tissues and organs and participate in different physiological and pathological molecular reactions in vivo. Wound healing is the basic process of traumatic disease recovery. Wound healing involves three overlapping stages: inflammation, proliferation, and remodelling. Furthermore, a comparison of wound healing with the tumour microenvironment revealed that galectin plays a key role in the wound healing process. The current review describes the role of galectin in inflammation, angiogenesis, re-epithelialisation, and fibrous scar formation and evaluates its potential as a therapeutic drug for wounds.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
https://doi.org/10.1007/s00109-022-02207-1
REVIEW
Regulation ofwound healing andfibrosis bygalectins
DongYu1,2· MingBu1,2· PingYu1· YapingLi1,2· YangChong1,2
Received: 2 December 2021 / Revised: 29 April 2022 / Accepted: 10 May 2022
© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2022
Abstract
Galectins are a family of proteins with at least one carbohydrate-recognition domain. Galectins are present in various tissues
and organs and participate in different physiological and pathological molecular reactions invivo. Wound healing is the basic
process of traumatic disease recovery. Wound healing involves three overlapping stages: inflammation, proliferation, and
remodelling. Furthermore, a comparison of wound healing with the tumour microenvironment revealed that galectin plays
a key role in the wound healing process. The current review describes the role of galectin in inflammation, angiogenesis,
re-epithelialisation, and fibrous scar formation and evaluates its potential as a therapeutic drug for wounds.
Keywords Galectin· Wound healing· Inflammation· Angiogenesis· Re-epithelialisation· Fibrosis
Introduction
Galectins are a family of proteins that binds to β-galactose
glucoside, all of which have one or two highly conserved
carbohydrate-recognition domains (CRDs) [1]. In total, 15
types of galectins have been discovered in mammals, of
which 11 exist in humans. They can be divided into three
groups according to their structures: (I) prototype galectin:
galectin-1 (Gal-1), -2, -5, -7, -10, -11, -13, -14, and -15,
which contain a dimerised CRD; (II) complex galectin: Gal-
4, -6, -8, -9, and -12, which contain two CRDs connected
by short peptides; and (III) chimeric galectin: Gal-3, with a
C-terminal CRD and an N-terminal tail [1, 2]. Galectins are
present in the cytoplasm and nucleus; they can be secreted
out of cells through various mechanisms [3]. Galectins can
bind to bacteria and agglutinate, and some galectins (Gal-4
and -8) can kill bacteria directly without activating other
factors such as complement factors [4, 5].
Galectins were first studied in-depth in tumour-related
fields. Galectins are key participants in mediating the inter-
action between tumour cells and host endothelial cells,
immune cells, and extracellular matrix (ECM), and they
participate in the regulation of tumour development and
progression, such as fibroblast activation, angiogenesis,
and immune response [6, 7]. Galectin has a high affinity
for glycans containing galactose glucoside on cell surfaces
and ECM glycoproteins [8, 9], which indicates its potential
involvement in wound healing. Some advances have been
made in studying the role of galectins in wound healing,
such as inflammation regulation, macrophage polarisation,
angiogenesis, fibroblast to myofibroblast transformation,
and re-epithelialisation [1, 1012]. The regulatory role
of galectins in tumour and wound healing makes them an
active candidate for treating cancer and skin wounds. The
present article reviews the role of galectins in various stages
of wound healing and discusses their potential and develop-
ment prospects in skin wound treatment (Table1).
Role ofgalectin inskin wound inflammation
Most galectins participate in the inflammation process in
the body. Gal-1 has an anti-inflammatory effect, and its
messenger RNA (mRNA) is up-regulated in the placenta
of women with preeclampsia, which is considered a foetal
response to maternal systemic infection [15]. Therapeutic
administration of Gal-1 can treat chronic T cell-dependent
inflammation, such as hepatitis, arthritis, and colitis [5355].
A subcutaneous injection of Gal-1 can accelerate the heal-
ing of pathological wounds in diabetic mice [20]. In addi-
tion, Gal-1 can stimulate mitogen-activated protein kinase
* Yang Chong
092016@yzu.edu.cn
1 Department ofTraditional Chinese Medicine, The Affiliated
Hospital ofYangzhou University, Yangzhou University,
Yangzhou225000, Jiangsu, China
2 Department ofMedical College, Yangzhou University,
Yangzhou225000, Jiangsu, China
/ Published online: 19 May 2022
Journal of Molecular Medicine (2022) 100:861–874
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... This unique feature allows Gal-4 to serve as a crucial crosslinker and regulator in numerous biological processes 6 . Research highlights its significant roles in stabilizing lipid rafts 8 , facilitating protein apical trafficking 22 , participating in wound healing 23 , influencing intestinal inflammation 24 , and contributing to tumour progression 25 , among other functions. The last decade, Gal-4 has also been implicated in HF 10 and diabetes [11][12][13][14][15] . ...
... There is limited research on whether Gal-4 specifically is involved in fibrosis. However, as indicated in a review by Yu et al., it is believed that galectins have similar and significant roles in the fibrotic process by promoting myofibroblasts to secrete extracellular matrix 23 . This is particularly pertinent, as fibrotic processes are often engaged to replace damaged myocardial tissue in IHD. ...
Article
Full-text available
An association between high Galectin-4 (Gal-4) and prevalence of diabetes in subjects with heart failure (HF) has previously been reported. The purpose of this study was to confirm these findings, as well as to further investigate this association, in a Swedish HF population. In addition, a second aim was to explore Gal-4’s association with obesity and biomarkers of metabolism and heart failure. Gal-4 was measured using a proximity extension array technique in 324 hospitalized HF patients within the Swedish HeArt and bRain failure investigation trial cohort. Obesity was defined as BMI ≥ 30. Multivariable logistic regression models were used to explore associations between Gal-4 and diabetes/obesity, and linear regression models were used to explore the associations between Gal-4 and biomarkers. A total of 309 participants (29.1% female; mean age 74.8 years) provided complete data for the analysis of associations between Gal-4 and diabetes. Additionally, for the analysis of heart failure phenotype, complete data was available for 230 subjects. Gal-4 was positively associated with prevalent diabetes (OR 2.60; CI 95% 1.56–4.32). In multivariable models, Gal-4 levels were significantly associated with obesity, but only for subjects with diabetes (OR 2.48; 1.09–5.62). Additionally, Gal-4 demonstrated a significant association with the incretin Glucose-dependent insulinotropic polypeptide (GIP), as well as with biomarkers of HF. In the stratified analyses, the association between Gal-4 and diabetes was prominent in patients with reduced ejection fraction (n = 160, OR 3.26; 95%CI 1.88–5.66), while it was not observed in those without (n = 70, 1.96 (0.75–5.10)). In this cross-sectional, observational study, higher Gal-4 levels in HF patients were associated with higher GIP levels. Further, increased levels of Gal-4 were associated with increased likelihood of diabetes, and obesity. This association was particularly pronounced in individuals with HF characterized by reduced ejection fraction. Additionally, Gal-4 levels were significantly elevated in heart failure patients with diabetes and obesity.
... As previously reported, multiple factors can contribute to poor wound healing in diabetes including reduced fibroblast proliferation and migration, and excessive fibroblast apoptosis (Hassanshahi et al. 2019;Yu et al. 2022). The function of fibroblast is to change tissue tension and is primarily responsible for wound healing or tissue contraction in the extracellular matrix of fibrotic organs Roman 2023). ...
Article
Full-text available
Slow wound healing in diabetic patients is a common complication of diabetes. Autologous conditioned serum (ACS) therapy is an emerging and safe biological therapy, and may accelerate the wound healing in diabetes. To investigate the effect of ACS in promoting wound healing in diabetic mice and its possible mechanism. Twenty-four six-week-old male C57BL/6 J mice were selected and divided into 5 groups, including control group (Ctrl), diabetic wound group (DW), ACS treatment group (DW+ACS) and STING pathway validation group (DW+ACS+DMXAA), with six mice in each group. Intervention was initiated after the back incision was performed, and wound healing was assessed on day 0, day 7, and day 14, and wound healing was assessed by hematoxylin and eosin (HE) staining of skin tissue on day 14. At the same time, the wound healing of the fibroblast markers collagen I and α-SMA was measured by immunohistochemistry and western blot. ACS treatment significantly accelerated the diabetic wound according to the wound area and HE staining results. Meanwhile, collagen I and α-SMA concentration evaluated by immunohistochemistry and western blot were remarkably elevated under the ACS interference. The STING signaling pathway was obviously activated in diabetic wound tissues. After the addition of DMXAA, an agonist of STING, the healing function of ACS was dramatically reversed. The application of ACS promotes wound healing in diabetic mice by enhancing fibroblasts. Meanwhile, the STING signaling pathway was inactivated by ACS interference. Hence, ACS can be used in the treatment of wound healing of Diabetes.
Article
Introduction: The rising incidence of liver diseases is a worldwide healthcare concern. However, the therapeutic options to manage chronic inflammation and fibrosis, the processes at the basis of morbidity and mortality of liver diseases, are very limited. Galectin 3 (Gal-3) is a protein implicated in fibrosis in multiple organs. Several Gal-3 inhibitors are currently in clinical development. Areas covered: This review describes our current understanding of the role of Gal-3 in chronic liver diseases, with special emphasis on fibrosis. Also, we review therapeutic advances based on Gal-3 inhibition, describing drug properties and their current status in clinical research. Expert opinion: Currently, the known effects of Gal-3 point to a direct activation of the NLRP3 inflammasome leading to its activation in liver macrophages and activated macrophages play a key role in tissue fibrogenesis. However, more research is needed to elucidate the role of Gal-3 in the different activation pathways, dissecting the intracellular and extracellular mechanisms of Gal-3, and its role in pathogenesis. Gal-3 could be a target for early therapy of numerous hepatic diseases and, given the lack of therapeutic options for liver fibrosis, there is a strong pharmacologic potential for Gal-3-based therapies.
Article
Full-text available
Lectin, a non-immunogenic origin protein, has properties of binding carbohydrates for the effective wound healing process. The process of wound healing is intricate and encompasses a diverse range of cells, extracellular matrix and soluble mediators involving the hemostasis, coagulation, inflammatory, proliferative, and remodeling or re-epithelialization phases. Lectin has shown to improve the infiltration of neutrophils, stimulate immune cells and regulate mediators like cytokine and growth factors, and promote angiogenesis. It also boosts the fibroblast synthesis, its differentiation into myofibroblast, and collagen deposition leading to complete re-epithelialization. Due to involvement of lectin in all these critical stages of wound healing results in complete wound closure. This review focuses on the role of lectin in wound healing and its potential application in wound-targeted formulations. It also covers its classification and historical background in a comprehensive manner.
Article
Full-text available
Background Tubulointerstitial fibrosis (TIF) is one of the main pathological features of various progressive renal damages and chronic kidney diseases. Mesenchymal stromal cells (MSCs) have been verified with significant improvement in the therapy of fibrosis diseases, but the mechanism is still unclear. We attempted to explore the new mechanism and therapeutic target of MSCs against renal fibrosis based on renal proteomics. Methods TIF model was induced by adenine gavage. Bone marrow-derived MSCs was injected by tail vein after modeling. Renal function and fibrosis related parameters were assessed by Masson, Sirius red, immunohistochemistry, and western blot. Renal proteomics was analyzed using iTRAQ-based mass spectrometry. Further possible mechanism was explored by transfected galectin-3 gene for knockdown (Gal-3 KD) and overexpression (Gal-3 OE) in HK-2 cells with lentiviral vector. Results MSCs treatment clearly decreased the expression of α-SMA, collagen type I, II, III, TGF-β1, Kim-1, p-Smad2/3, IL-6, IL-1β, and TNFα compared with model rats, while p38 MAPK increased. Proteomics showed that only 40 proteins exhibited significant differences (30 upregulated, 10 downregulated) compared MSCs group with the model group. Galectin-3 was downregulated significantly in renal tissues and TGF-β1-induced rat tubular epithelial cells and interstitial fibroblasts, consistent with the iTRAQ results. Gal-3 KD notably inhibited the expression of p-Akt, p-GSK3β and snail in TGF-β1-induced HK-2 cells fibrosis. On the contrary, Gal-3 OE obviously increased the expression of p-Akt, p-GSK3β and snail. Conclusion The mechanism of MSCs anti-renal fibrosis was probably mediated by galectin-3/Akt/GSK3β/Snail signaling pathway. Galectin-3 may be a valuable target for treating renal fibrosis.
Article
Full-text available
Within this work, a new class of sequence‐defined heteromultivalent glycomacromolecules bearing lactose residues and nonglycosidic motifs for probing glycoconjugate recognition in carbohydrate recognition domain (CRD) of galectin‐3 is presented. Galectins, a family of β‐galactoside‐binding proteins, are known to play crucial roles in different signaling pathways involved in tumor biology. Thus, research has focused on the design and synthesis of galectin‐targeting ligands for use as diagnostic markers or potential therapeutics. Heteromultivalent precision glycomacromolecules have the potential to serve as ligands for galectins. In this work, multivalency and the introduction of nonglycosidic motifs bearing either neutral, amine, or sulfonated/sulfated groups are used to better understand binding in the galectin‐3 CRD. Enzyme‐linked immunosorbent assays and surface plasmon resonance studies are performed, revealing a positive impact of the sulfonated/sulfated nonglycosidic motifs on galectin‐3 binding but not on galectin‐1 binding. Selected compounds are then tested with galectin‐3 positive MCF 7 breast cancer cells using an in vitro would scratch assay. Preliminary results demonstrate a differential biological effect on MCF 7 cells with high galectin‐3 expression in comparison to an HEK 293 control with low galectin‐3 expression, indicating the potential for sulfonated/sulfated heteromultivalent glycomacromolecules to serve as preferential ligands for galectin‐3 targeting.
Article
Full-text available
Fibrosis is the extensive deposition of fibrous connective tissue, and it is characterized by the accumulation of collagen and other extracellular matrix (ECM) components. Fibrosis is essential for wound healing and tissue repair in response to a variety of triggers, which include infection, inflammation, autoimmune disorder, degenerative disease, tumor, and injury. Fibrotic remodeling in various diseases, such as liver cirrhosis, pulmonary fibrosis, renal interstitial fibrosis, myocardial infarction, systemic sclerosis (SSc), and graft-versus-host disease (GVHD), can impair organ function, causing high morbidity and mortality. Both innate and adaptive immunity are involved in fibrogenesis. Although the roles of macrophages in fibrogenesis have been studied for many years, the underlying mechanisms concerning the manner in which T cells regulate fibrosis are not completely understood. The T cell receptor (TCR) engages the antigen and shapes the repertoire of antigen-specific T cells. Based on the divergent expression of surface molecules and cell functions, T cells are subdivided into natural killer T (NKT) cells, γδ T cells, CD8⁺ cytotoxic T lymphocytes (CTL), regulatory T (Treg) cells, T follicular regulatory (Tfr) cells, and T helper cells, including Th1, Th2, Th9, Th17, Th22, and T follicular helper (Tfh) cells. In this review, we summarize the pro-fibrotic or anti-fibrotic roles and distinct mechanisms of different T cell subsets. On reviewing the literature, we conclude that the T cell regulations are commonly disease-specific and tissue-specific. Finally, we provide perspectives on microbiota, viral infection, and metabolism, and discuss the current advancements of technologies for identifying novel targets and developing immunotherapies for intervention in fibrosis and fibrotic diseases.
Article
Full-text available
The expression of prototype galectin‐14 (Gal‐14) in human placenta is higher than any other galectin, suggesting that it may play a role in fetal development and regulation of immune tolerance during pregnancy. Here, we solved the crystal structure of dimeric Gal‐14 and found that its global fold is significantly different from that of other galectins with two β‐strands (S5 and S6) extending from one monomer and contributing to the carbohydrate‐binding domain of the other. The hemagglutination assay showed that this lectin could induce agglutination of chicken erythrocytes, even though lactose could not inhibit Gal‐14‐induced agglutination activity. Calorimetry indicates that lactose does not interact with this lectin. Compared to galectin‐1, galectin‐3, and galectin‐8, Gal‐14 has two key amino acids (a histidine and an arginine) in the normally conserved, canonical sugar‐binding site, which are substituted by glutamine (Gln53) and histidine (His57), thus likely explaining why lactose binding to this lectin is very weak. Lactose was observed in the ligand‐binding site of one Gal‐14 structure, most likely because ligand binding is weak and crystals were allowed to grow over a long period of time in the presence of lactose. We also found that EGFP‐tagged Gal‐14 is primarily localized within the nucleus of different cell types. In addition, Gal‐14 colocalized with c‐Rel (a member of NF‐κB family) in HeLa cells. These findings indicate that Gal‐14 might regulate signal transduction pathways through NF‐κB hubs. Overall, the present study provides impetus for further research into the function of Gal‐14 in embryology.
Article
Full-text available
Background and Objective During cyclosporine‐induced gingival overgrowth, the homeostatic balance of gingival connective tissue is disrupted leading to fibrosis. Galectins are glycan‐binding proteins that can modulate a variety of cellular processes including fibrosis in several organs. Here, we study the role of galectin‐8 (Gal‐8) in the response of gingival connective tissue cells to cyclosporine. Methods We used human gingival fibroblasts and mouse NIH3T3 cells treated with recombinant Gal‐8 and/or cyclosporine for analyzing specific mRNA and protein levels through immunoblot, real‐time polymerase chain reaction, ELISA and immunofluorescence, pull‐down with Gal‐8‐Sepharose for Gal‐8‐to‐cell surface glycoprotein interactions, short hairpin RNA for Gal‐8 silencing and Student's t test and ANOVA for statistical analysis. Results Galectin‐8 stimulated type I collagen and fibronectin protein levels and potentiated CTGF protein levels in TGF‐β1‐stimulated human gingival fibroblasts. Gal‐8 interacted with α5β1‐integrin and type II TGF‐β receptor. Gal‐8 stimulated fibronectin protein and mRNA levels, and this response was dependent on FAK activity but not Smad2/3 signaling. Cyclosporine and tumor necrosis factor alpha (TNF‐α) increased Gal‐8 protein levels. Finally, silencing of galectin‐8 in NIH3T3 cells abolished cyclosporine‐induced fibronectin protein levels. Conclusion Taken together, these results reveal for the first time Gal‐8 as a fibrogenic stimulus exerted through β1‐integrin/FAK pathways in human gingival fibroblasts, which can be triggered by cyclosporine. Further studies should explore the involvement of Gal‐8 in human gingival tissues and its role in drug‐induced gingival overgrowth.
Article
Full-text available
K-Ras is the most prominent driver of oncogenesis and no effective K-Ras inhibitors have been established despite decades of intensive research. Identifying new K-Ras-binding proteins and their interaction domains offers the opportunity for defining new approaches in tackling oncogenic K-Ras. We have identified Galectin-8 as a novel, direct binding protein for K-Ras4B by mass spectrometry analyses and protein interaction studies. Galectin-8 is a tandem-repeat Galectin and it is widely expressed in lung and pancreatic carcinoma cells. siRNA-mediated depletion of Galectin-8 resulted in increased K-Ras4B content and ERK1/2 activity in lung and pancreatic carcinoma cells. Moreover, cell migration and cell proliferation were inhibited by the depletion of Galectin-8. The K-Ras4B-Galectin-8 interaction is indispensably associated with the farnesylation of K-Ras4B. The lysine-rich polybasic domain (PBD), a region that is unique for K-Ras4B as compared to H- and N-Ras, stabilizes the interaction and accounts for the specificity. Binding assays with the deletion mutants of Galectin-8, comprising either of the two carbohydrate recognition domains (CRD), revealed that K-Ras4B only interacts with the N-CRD, but not with the C-CRD. Structural modeling uncovers a potential binding pocket for the hydrophobic farnesyl chain of K-Ras4B and a cluster of negatively charged amino acids for interaction with the positively charged lysine residues in the N-CRD. Our results demonstrate that Galectin-8 is a new binding partner for K-Ras4B and it interacts via the N-CRD with the farnesylated PBD of K-Ras, thereby modulating the K-Ras effector pathways as well as cell proliferation and migration.
Article
Hyperactive angiogenesis contributes to the immunosuppressive microenvironment important for immunotherapy. Galectin‐1, encoded by LGALS1, can trigger the vascular signaling programs and mediate the anti‐angiogenic treatment response. However, the mechanism through which galectin‐1 regulates angiogenesis is poorly understood. It has been suggested that galectin‐1 may associate with mRNAs in cells. This study applied the iRIP‐seq methodology to study the potential role of galectin‐1 as an RNA‐binding protein. We found that galectin‐1 interacts with a large number of mRNAs, with a preference for binding near stop codons and a preference for UGCA/UGGA and GAGCAG as binding motifs. Galectin‐1 binds to the mRNAs of angiogenesis‐associated genes including VEGFA, EGR1 and LAMA5, suggesting that galectin‐1 may regulate angiogenesis via its mRNA‐binding activity. We further show that shLGALS1 inhibits capillary tube formation in an in vitro angiogenesis assay and alters the expression levels of several galectin‐1‐bound angiogenesis‐associated mRNAs. These results uncover a previously unrecognized mRNA‐binding activity of galectin‐1.
Article
Upon healing, burn wounds often leave hypertrophic scars (HTSs) marked by excess collagen deposition, dermal and epidermal thickening, hypervascularity, and an increased density of fibroblasts. The Galectins, a family of lectins with a conserved carbohydrate recognition domain, function intracellularly and extracellularly to mediate a multitude of biological processes including inflammatory responses, angiogenesis, cell migration and differentiation, and cell‐ECM adhesion. Galectin‐1 (Gal‐1) has been associated with several fibrotic diseases and can induce keratinocyte and fibroblast proliferation, migration, and differentiation into fibroproliferative myofibroblasts. In this study, Gal‐1 expression was assessed in human and porcine HTS. In a microarray, galectins 1, 4, and 12 were upregulated in pig HTS compared to normal skin (fold change = +3.58, +6.11, and +3.03, FDR <0.01). Confirmatory qRT‐PCR demonstrated significant upregulation of Galectin‐1 (LGALS1) transcription in HTS in both human and porcine tissues (fold change = +7.78 and +7.90, P <.05). In pig HTS, this upregulation was maintained throughout scar development and remodeling. Immunofluorescent staining of Gal‐1 in human and porcine HTS showed significantly increased fluorescence (202.5 ± 58.2 vs 35.2 ± 21.0, P <.05 and 276.1 ± 12.7 vs 69.7 ± 25.9, P <.01) compared to normal skin and co‐localization with smooth muscle actin‐expressing myofibroblasts. A strong positive correlation (R = .948) was observed between LGALS1 and Collagen type 1 alpha 1 mRNA expression. Gal‐1 is overexpressed in HTS at the mRNA and protein levels and may have a role in the development of scar phenotypes due to fibroblast over‐proliferation, collagen secretion, and dermal thickening. The role of galectins shows promise for future study and may lead to the development of a pharmacotherapy for treatment of HTS.
Article
Psoriasis is a chronic inflammatory skin disease characterized by inflammatory cell infiltration, as well as hyperproliferation of keratinocytes in skin lesions, and is considered a metabolic syndrome. We found that the expression of galectin-7 is reduced in the skin lesions of patients with psoriasis. IL-17A and TNF-α, two cytokines intimately involved in the development of psoriatic lesions, suppressed galectin-7 expression in human primary keratinocytes (HEKn cells) and the immortalized human keratinocyte cell line HaCaT. A galectin-7 knockdown in these cells elevated the production of IL-6 and IL-8 and enhanced ERK signaling when the cells were stimulated with IL-17A. Galectin-7 attenuated IL-17A-induced production of inflammatory mediators by keratinocytes via the miR-146a-ERK pathway. Moreover, galectin-7-deficient mice showed enhanced epidermal hyperplasia and skin inflammation in response to intradermal IL-23 injection. We identified fluvastatin as an inducer of galectin-7 expression by connectivity map (cMAP) analysis, confirmed this effect in keratinocytes, and demonstrated that fluvastatin attenuated IL-6 and IL-8 production induced by IL-17A. Thus, we validate a role of galectin-7 in the pathogenesis of psoriasis, in both epidermal hyperplasia and keratinocyte-mediated inflammatory responses, and formulated a rationale for the use of statins in the treatment of psoriasis.
Article
Galectin-4 is a member of multifunctional galactoside-binding lectin family with various biological functions, including tumor cells proliferation, cancer progression, cell adhesion, and tumor metastasis. In this study, we aimed to investigate the putative function of galectin-4 in tumor progression of hepatocellular carcinoma (HCC), and explore the possible mechanisms of galectin-4 related biological pathway. The result showed that the galectin-4 was significantly up-regulated in HCC tissues/cell lines compared to matched peritumor tissues/a normal liver cell line, respectively. Furthermore, overexpressing galectin-4 in HCCLM3 cells led to promotion of cell proliferation and inhibition of cell apoptosis via up-regulation of Cyclin D1, down-regulation of p21 and decreased pro-apoptotic Bax/Bcl-2 ratio, But knockdown of galectin-4 reversed these properties. Furthermore, galectin-4 could promote cell-cell adhesion via stabilizing the adherens junction complexes. These observations was further validated in the xenograft animal model. Taken together, galectin-4 plays an important role in HCC progression.