Next Article in Journal
Engineering Innate Immunity: Recent Advances and Future Directions for CAR-NK and CAR–Macrophage Therapies in Solid Tumors
Previous Article in Journal
Anticancer Effect of Nature-Inspired Indolizine-Based Pentathiepines in 2D and 3D Cellular Model
Previous Article in Special Issue
The Role of Pulmonary Collectins, Surfactant Protein A (SP-A) and Surfactant Protein D (SP-D) in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

REG3A: A Multifunctional Antioxidant Lectin at the Crossroads of Microbiota Regulation, Inflammation, and Cancer

1
National Institute of Health and Medical Research INSERM, UMR 1193, Paris-Saclay University, Paul-Brousse University Hospital, Hepatobiliary Centre, 94800 Villejuif, France
2
Medical School, Paris-Saclay University, 94270 Le Kremlin-Bicêtre, France
3
Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Saclay, Hospital-University Department of Biology and Genetics, Paul-Brousse Hospital, 94800 Villejuif, France
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(14), 2395; https://doi.org/10.3390/cancers17142395
Submission received: 22 May 2025 / Revised: 16 July 2025 / Accepted: 16 July 2025 / Published: 19 July 2025
(This article belongs to the Special Issue Lectins in Cancer)

Simple Summary

REG3A, a key member of the human REG lectin family, plays a multifaceted role in immunity, inflammation, and cancer. Primarily expressed in gastrointestinal epithelial cells, it reinforces gut barrier integrity, preserves mucosal immunity, and regulates host–microbiota interactions. Beyond its antimicrobial functions, REG3A acts as a targeted non-enzymatic antioxidant, protecting tissues from oxidative damage. Its expression is tightly regulated by inflammatory signals and is markedly upregulated during immune activation, where it limits microbial invasion, reduces tissue injury, and promotes repair. While REG3A offers critical protection in inflammatory settings, its role in cancer is far more complex. Depending on the tumor type and microenvironment, REG3A can either support tumor growth or exert tumor-suppressive effects. This review highlights the multifaceted biology of REG3A, with a focus on its roles in epithelial defense, immune modulation, oxidative stress regulation, and its paradoxical functions in cancer.

Abstract

REG3A, a prominent member of the human regenerating islet-derived (REG) lectin family, plays a pivotal and multifaceted role in immune defense, inflammation, and cancer biology. Primarily expressed in gastrointestinal epithelial cells, REG3A reinforces barrier integrity, orchestrates mucosal immune responses, and regulates host–microbiota interactions. It also functions as a potent non-enzymatic antioxidant, protecting tissues from oxidative stress. REG3A expression is tightly regulated by inflammatory stimuli and is robustly induced during immune activation, where it limits microbial invasion, dampens tissue injury, and promotes epithelial repair. Beyond its antimicrobial and immunomodulatory properties, REG3A contributes to the resolution of inflammation and the maintenance of tissue homeostasis. However, its role in cancer is highly context-dependent. In some tumor types, REG3A fosters malignant progression by enhancing cell survival, proliferation, and invasiveness. In others, it acts as a tumor suppressor, inhibiting growth and metastatic potential. These opposing effects are likely dictated by a combination of factors, including the tissue of origin, the composition and dynamics of the tumor microenvironment, and the stage of disease progression. Additionally, the secreted nature of REG3A implies both local and systemic effects, further modulated by organ-specific physiology. Experimental variability may also reflect differences in methodologies, analytical tools, and model systems used. This review synthesizes current knowledge on the pleiotropic functions of REG3A, emphasizing its roles in epithelial defense, immune regulation, redox homeostasis, and oncogenesis. A deeper understanding of REG3A’s pleiotropic effects could open up new therapeutic avenues in both inflammatory disorders and cancer.

1. Introduction

The REG3 proteins, members of the regenerating islet-derived (REG) lectin family, were first identified in 1984 following their isolation from rat pancreatic islets [1]. Among them, REG3A has garnered particular interest due to its multifunctional roles in tissue regeneration, immune regulation, and host defense. Over time, REG3A has been designated by several names depending on the species and physiological conditions in which it was studied. It was initially described as Pancreatitis-Associated Protein (PAP or PAP1) after its identification in a rat model of acute pancreatitis [2], and as Hepatocarcinoma-Intestine-Pancreas protein (HIP) in human hepatocellular carcinoma [3]. Other less commonly used aliases have included peptide 23, Islet Neogenesis-Associated Protein (INGAP), and Pancreatic Thread Protein (PTP). While the term HIP/PAP persisted in the early literature, the protein is now widely referred to as REG3A, in line with its classification within the REG family of C-type lectins with regenerative properties. Notably, some studies have incorrectly referred to REG3A as REG2, which is in fact a distinct murine-specific family member without a human homolog [4].
The REG protein family is subdivided into four groups: REG1, REG2, REG3, and REG4. Except for REG4, which is located on chromosome 1, all other REG genes in humans are clustered on chromosome 2p12 (corresponding to chromosome 16p12–13 in mice) [5], reflecting their derivation from a common ancestral gene and subsequent duplication during evolution. This genetic clustering implies both functional conservation and potential redundancy among family members. The REG1 subgroup comprises REG1A and REG1B in human [6,7] and mice, while the REG3 subgroup includes REG3A and REG3G in humans, and Reg3α, Reg3β, Reg3δ, and Reg3γ in mice. All REG3 members share significant sequence homology both within and across species [5]. Human REG3A shares 69.1% and 66.3% sequence identity—and 80.9% and 82.4% similarity—with mouse Reg3β and Reg3γ, respectively. All three proteins conserve the functional EPN lectin motif at the apex of loop 1. In loop 2, however, the ERN motif found in REG3A and Reg3β is substituted by an ETN motif in Reg3γ. Cysteine and aromatic residues are fully conserved across these orthologs. The primary sequence divergence lies in the carboxy-terminal region, for which no defined functional motif has been identified. Mouse Reg3α shows greater divergence, featuring a QPN motif in loop 1 and a DGD sequence in loop 2, indicating distinct lectin-binding specificity. Among these, Reg3β and Reg3γ are considered the murine orthologs of human REG3A, while Reg3α is orthologous to human REG3G. The diversity of nomenclature across species and the frequent use of different orthologs in experimental models have complicated the functional characterization of REG3A. For the sake of consistency, the term REG3A will be used throughout this article to refer to both the human protein and its murine counterparts Reg3β and Reg3γ.
REG3A is initially synthesized as a 175-amino acid prepropeptide (19 kDa) (Figure 1) [3,8,9]. Proteolytic removal of the 26-amino acid signal peptide results in the secretion of the mature ~16 kDa form into the intestinal lumen and, to a lesser extent, into the systemic circulation [9,10]. The N-terminal pro-segment keeps REG3A in an inactive state until cleaved by host or microbial proteases. Activation occurs through cleavage at specific sites by digestive enzymes, trypsin at Arg37–Ile38 and elastase at Ser35–Ala36, or by bacterial proteases such as Bacillus subtilis npr9 [11,12,13]. In vitro studies have shown that 1.5 µM REG3A is fully cleaved by 10 nM trypsin within 5 min, and by 50 nM and 150 nM of elastase 2A and 2B, respectively, within 30 min. Similarly, Bacillus subtilis culture supernatant can fully cleave 1.5 µM REG3A within the same time frame [13]. The bacterium-aggregating activities of the resulting cleavage products were evaluated and found to be comparable. Interestingly, trypsin cleavage generates a 15 kDa polymerizable form that assembles into insoluble, fibrillar structures, whereas elastase cleavage preserves REG3A in a soluble form [12,14].
The three-dimensional structure of REG3A has been resolved by X-ray crystallography (PDB ID: 1UV0 [10] and NMR spectroscopy (PDB ID: 2GO0) [11,15]). Its structure includes two α-helices, eight β-sheets, and three disulfide bridges (Cys 40-Cys51, Cys68-Cys171, Cys146-Cys163), forming a classical carbohydrate recognition domain (CRD) (Figure 1). REG3A belongs to subgroup VIII of C-type lectins, which are characterized by a single CRD that binds carbohydrates in a calcium-independent manner, typically through conserved motifs such as EPN (Glu–Pro–Asn), QPD (Gln–Pro–Asp), or QPN (Gln–Pro–Asn) [8,16,17]. REG3A uniquely positions its EPN motif on loop 1 (residues 107–121) and the ERN motif on loop 2 (residues 130–145), contributing to its distinct ligand specificity [18] (Figure 1).
Functionally, REG3A is well known for its lectin activity, particularly its ability to bind pathogen-associated molecular patterns (PAMPs) such as mannose and N-acetylglucosamine (GlcNAc) residues [19]. Its affinity increases significantly when these carbohydrates are part of polysaccharide structures like peptidoglycan, lipopolysaccharides (LPS), mannan, and chitin. This binding underlies its potent bactericidal activity against Gram-positive microorganisms, such as Enterococcus faecalis and Listeria spp., especially in its cleaved, active form within the intestinal lumen [19]. In addition to microbial recognition, REG3A can bind glucose and its phosphorylated derivatives involved in the hexosamine biosynthesis pathway, thereby influencing key metabolic processes such as protein O-GlcNAcylation, complex glycosylation, and glycolysis [20].
Beyond its lectin functions, REG3A has emerged as a potent reactive oxygen species (ROS) scavenger, with a strong affinity for hydroxyl radicals [21]. Accumulating evidence supports its role in protecting tissues against oxidative injury. In acute liver failure, REG3A binds to fibrin scaffolds in necrotic areas of the liver, where it reduces ROS levels and promotes hepatocyte survival [22]. In muscle cells, REG3A prevents oxidative damage to the glycoprotein GP130, thereby preserving GP130/AMPK signaling and enhancing glucose uptake [23]. However, the underlying antioxidant mechanisms remain incompletely understood, particularly whether REG3A engages with canonical redox pathways such as Nrf2 signaling. Given the pivotal role of Nrf2 in regulating antioxidant responses, it is plausible that REG3A may influence or interact with this pathway. Further studies are warranted to determine whether REG3A modulates Nrf2 activity, either directly or indirectly through its effect on oxidative stress, as well as its potential impact on other antioxidant mediators, including superoxide dismutase (SOD), catalase, and heme oxygenase-1 (HO-1).
In summary, REG3A exhibits a multifunctional profile combining lectin activity, antimicrobial properties, and ROS-scavenging capacity. It binds bacterial cell wall carbohydrates in a calcium-independent manner, disrupts bacterial membranes to exert bactericidal effects, or protects oxygen-sensitive commensals from oxidative damage. These diverse functions contribute to epithelial barrier defense, immune modulation, and the maintenance of tissue homeostasis. The following sections will examine the implications of REG3A’s dual functionality in inflammation, host–microbe interactions, and tumor biology.

2. REG3A: Expression and Regulation

REG3A is predominantly expressed in gastrointestinal organs such as the pancreas [24], stomach [25], and intestinal tract [26]. Its expression is highly inducible in response to infection and inflammation (Table 1). At lower levels, REG3A is also detected in a limited number of other cells and tissues, including cholangiocytes [27], skin [28], uterus [29], and the pituitary gland [30]. A key driver of REG3A expression in the gut is bacterial colonization [19,31]. In germ-free or antibiotic-treated animals, REG3A expression in the intestinal lumen is minimal, whereas probiotic supplementation significantly enhances its expression [19,31,32,33]. Paneth cells play a central role by sensing bacterial signals through the TLR-MyD88 signaling pathway, thereby promoting REG3A transcription [34]. In addition, IL-22, produced by innate lymphoid cells, further stimulates REG3A expression [35], thereby enhancing its role in maintaining intestinal barrier function and in protecting intestinal stem cells. REG3A plays a role in shaping the gut microbiome and maintaining spatial separation between bacteria and the intestinal epithelium [36].
REG3A expression is modulated under various pathophysiological conditions. It is robustly upregulated during bacterial infections, supporting its function as a protective factor against microbial invasion [37,38]. For instance, infections with Listeria monocytogenes and Salmonella enteritidis induce REG3A expression in the intestine, with corresponding effects on mucus distribution [39]. Furthermore, the promoter region of the REG3A gene contains a functional IL-6 responsive element, highlighting a close connection between REG3A regulation and the immune response [40]. Other cytokines including IL-8, IL-17, IL-22, IL-33, and IL-36, have also been shown to upregulate REG3A expression [41,42,43,44]. Overall, REG3A functions as an acute-phase protein and is a key effector in the innate immune response.
REG3A expression is markedly upregulated in response to both acute and chronic tissue stress, as well as across a range of malignancies. A comprehensive summary of these expression patterns is provided in Table 1 and Table 2. Owing to its strong diagnostic relevance, REG3A has gained recognition as a circulating biomarker in several pathological conditions, where elevated serum levels often correlate with disease severity and poor prognosis. In chronic colitis [45], gastrointestinal graft-versus-host disease (GI-GvHD) [46], sepsis [47], and pancreatic ductal adenocarcinoma (PDAC) [48], increased REG3A levels reflect heightened disease activity. Notably, in GI-GvHD, plasma REG3A concentrations are significantly higher in affected patients compared to those without gastrointestinal involvement. A threshold of ≥72 ng/mL has been shown to predict a 1.9-fold increased risk of non-relapse mortality, independent of other clinical variables, establishing REG3A as a validated prognostic marker in this setting [46,49]. In sepsis, REG3A has recently emerged as both a diagnostic and prognostic biomarker. Serum levels correlate strongly with clinical severity scores (SOFA and APACHE II), accurately distinguish septic patients from healthy controls, and independently predict 28-day survival [47]. Similarly, in rectal cancer patients undergoing neoadjuvant chemoradiotherapy (CCRT), transcriptomic analyses identified REG3A as the most upregulated gene in non-responders (log2 ≈ 1.25, p = 0.0079). High REG3A protein expression is associated with poor treatment response, increased lymph node metastasis, vascular invasion, and serves as an independent predictor of both disease-specific and metastasis-free survival [50]. Furthermore, REG3A shows promising utility as a urinary biomarker in bladder cancer. In a single-cohort study, REG3A demonstrated a sensitivity of 80.2%, specificity of 78.2%, positive predictive value of 75.7%, negative predictive value of 82.3%, and an area under the curve (AUC) of 0.863, outperforming conventional markers such as NMP22 (sensitivity 52.1%, specificity 93.5%) [51]. Collectively, these findings underscore the emerging clinical value of REG3A as a biomarker in both inflammatory and oncologic gastrointestinal diseases.
Table 1. REG3A expression patterns across tissues and clinical conditions.
Table 1. REG3A expression patterns across tissues and clinical conditions.
OrgansCondition
PhysiologicalInflammatoryMalignant
Pancreasβ-cells (islets)Pancreatitis [24],
(inflamed acini)
Cystic fibrosis [52]
Adenocarcinoma [53]
StomachEpithelial cellsN/AGastric cancer [54]
Small intestinePaneth cellsPaneth cells
Enterocytes
ColonNot expressedColitis [55], Celiac disease, Crohn’s disease [56] (enterocytes) [55]Colorectal cancer [57]
LiverCholangiocytesHepatitis [21]
(inflamed hepatocytes) [58,59]
Hepatocellular carcinoma [3,60]
Cholangiocarcinoma [27]
BladderNot expressedN/ABladder cancer
SkinKeratinocytesSkin lesions
Psoriasis [28]
Mammary glandN/AN/ABreast cancer
BrainMotoneurons [61]Brain injury [62]
Alzheimer [63]
Pituitary glandGrowth hormone producing cellsN/AAdenoma [64]
LungN/AFibrosis [65]Lung cancer
TracheaN/AN/ASquamous cell carcinoma [66]
PBMCsN/ALupus nephritis [67],
Polymyositis
Dermatomyositis [68]
Gastric cancer [54]
Abbreviations: N/A: not available.
Table 2. Comparative analysis of REG3A studies across cancer types and experimental models (in vitro, in vivo, human cohorts).
Table 2. Comparative analysis of REG3A studies across cancer types and experimental models (in vitro, in vivo, human cohorts).
Link With CancerConditions
In Vitro In VivoHuman Cohort
Colorectal
cancer
SuppressorBMI1/MEL18 repress REG3A transcription [69]

REG3A binds
IL6/GP130 [69]

REG3A inhibits IL6/STAT3 signaling [69]
rREG3A reduces polyps in AOM-DSS model [69]REG3A inversely correlates
with cancer (331 pairs NT/T) [57]

High REG3A associates
with better OS (n = 279) [57,69]

REG3A inversely correlates
with venous invasion [57]

REG3A inversely correlates
with STAT3 activation [69]
PromoterREG3A induces AKT
and ERK [70]
REG3A promotes
tumor growth
in xenografts [70]
REG3A positively correlates
with cancer (n = 335) [70]

High REG3A associates with
tumor size, differentiation,
stage (n = 82) [70]
Gastric
cancer
SuppressorREG3A inhibits AKT
and GSK3β activation.

REG3A induces
apoptosis.

REG3A reduces
cell invasion [71]
N/AREG3A negatively correlates with cancer (30 NT/T pairs [25]; n = 876 [71])

High REG3A associates with better OS (n = 311) and disease-free survival (n = 565) [71]

Promoter
REG3A inhibits
proliferation, migration and invasion [72]

REG3A suppresses JAK2/STAT3 signaling [72]
N/AREG3A upregulated in cancer
(41 NT/T pairs) [72]
Hepato-
cellular
carcinoma
SuppressorWnt/β-catenin
induces REG3A [60]

REG3A reduces
proliferation [20]

REG3A binds
hexosamine pathway sugars [20]

REG3A lowers
O-GlcNAcylation [20]
REG3A inhibits
cancer development (WHC/MYC, DEN) [20]

REG3A lowers
UDP-GlcNAc [20]
REG3A localizes to cirrhotic
hepatocytes [26,27,58]

REG3A links to low stage HCC, no vascular invasion, β-catenin mutation (n = 265) [73]

REG3A associates with
β-catenin activation (42 NT/T) [60]

REG3A improves disease-free
survival in cirrhosis (n = 216) [20]

No link between REG3A and OS
in patients with HCC (n = 507) [20]
PromoterHSC/HCC coculture
induces REG3A via PDGFββ,
reducing apoptosis [74]

REG3A knockdown
in coculture lowers p42//44 activation [74]
REG3A reduces
tumor growth of Lx2/MH134 xenografts in C3H mice [74]
REG3A positively correlates
with cancer (88 NT/T pairs) [74]
Pancreatic cancerSuppressorN/AN/AN/A
PromoterREG3A promotes
proliferation, migration, and invasion;
activates EGFR and GP130/JAK2/STAT3 [48,75,76,77]

IL6 induces REG3A via JAK2/STAT3 [75]

REG3A inhibits DC
maturation [78]

REG3A drives acini-to-ductal metaplasia via EXTL3/RAS/ERK
(3D culture) [79]
REG3A promotes
cancer in
DMBA-caerulein and
Pdx1-Cre; KrasG12D; Ink4a/Arffl/fl models [48,76,79,80]

REG3A enhances
tumor growth and
induces immunosuppression in Panc02 xenografts [78]

REG3A activates EGFR/JAK2/STAT3 and synergizes with IL6 to boost tumors in xenograft mice [75,78]
Serum REG3A < 17.5 µg/L links to cancer (n = 254) [48], high TNM grade (n = 58) [77] and poor survival [48,77]

REG3A localizes to peritumoral acini and CK19+ preneoplastic lesions [48,77,79]

REG3A correlates with IL6/JAK/STAT3 in peritumoral acini [77]
Abbreviations: rREG3A: recombinant REG3A; OS: overall survival; HCC: hepatocellular carcinoma; HSC: hepatic stellate cell; DC: dendritic cell; N/A: not available.

3. REG3A: Balancing Defense and Harmony in the Gut

REG3A plays a central role in intestinal antimicrobial defense, with a preference for targeting Gram-positive bacteria. Cash et al. showed that its luminal, truncated form exerts potent bactericidal activity against organisms such as Enterococcus faecalis and various Listeria strains [19]. This effect is mediated through the direct binding of REG3A to bacterial peptidoglycan, which anchors the protein to the bacterial surface. Upon binding, REG3A facilitates pore formation in the membrane, leading to cytoplasmic leakage and bacterial cell death [11,81] (Figure 2). Through this mechanism, REG3A functions as an antimicrobial protein (AMP), cooperating with other intestinal AMPs to reinforce the epithelial barrier and maintain gut homeostasis [82]. Interestingly, REG3A also demonstrates bactericidal activity against select Gram-negative bacteria, including Clostridium butyricum, Lactobacillus reuteri, and several Escherichia coli strains [83]. This activity is thought to be mediated via binding to LPS, the hallmark component of Gram-negative outer membranes [84] (Figure 2). By exerting these antimicrobial effects, REG3A contributes to maintain spatial segregation between gut microbes and the epithelium surface [36], a critical defense that limits bacterial translocation to extra-intestinal sites, such as the liver [85].
Although a number of Gram-positive and Gram-negative species have been classified as either susceptible or resistant to REG3A, the molecular basis for this selectivity remains incompletely understood. Studies using Salmonella Typhimurium mutants with modified LPS structures suggest that REG3A’s activity may depend on its ability to recognize specific peptidoglycan and LPS configurations [83]. Understanding these interactions is further complicated by interspecies differences: whereas humans express a single REG3A isoform, mice produce two orthologs, REG3α and REG3γ, which may differ in their bacterial specificity. The role of REG3A in host–microbe interactions extends far beyond pathogen clearance (Figure 2). Rather than indiscriminately clearing bacteria, a strategy that could be evolutionarily detrimental, REG3A promotes intestinal immune homeostasis by sculpting a balanced and protective microbial ecosystem. Compelling evidence revealed that REG3A shapes gut microbiota composition, enriching Clostridiales, while depleting Bacteroidetes and Proteobacteria. This microbial shift correlated with reduced oxidative stress and intestinal inflammation, leading to a lower susceptibility to colitis (Figure 2). Notably, REG3A was shown to decrease ROS levels in the gut microbiota of mice with colitis and enhance the survival of oxygen-sensitive, Gram-positive Clostridia in vitro, suggesting that its antioxidant activity contributes to microbiota modulation [86].
Animal studies highlight the diet-sensitive nature of REG3A expression and its influence on gut microbial ecology. Frazier et al. [87] showed that REG3A expression in the intestinal epithelium follows a diurnal rhythm, paralleling oscillations in Lactobacillus abundance and suppression of Peptostreptococcaceae. A high-fat diet disrupts this pattern, flattening REG3A expression, reducing Lactobacillus fluctuations, and promoting Clostridiaceae and Peptostreptococcaceae expansion, microbial shifts linked to impaired glucose regulation and metabolic dysfunction. In contrast, fermentable fibers such as inulin markedly upregulate REG3A expression. Shin et al. [33] demonstrated that this fiber-induced REG3A expression is crucial for the metabolic benefits of dietary fiber, including improved glucose tolerance and the expansion of beneficial microbes like Bifidobacteria. Together, these findings establish REG3A as a key mediator linking diet, microbial dynamics, and host metabolic health.

4. REG3A in Innate Immunity: Sensing DAMPs and Modulating Anti-Inflammatory Responses

While REG3A is well-known for recognizing microbial sugars, it also binds to damage-associated molecular patterns (DAMPs), particularly within the inflammatory extracellular matrix (ECM) during acute hepatitis. In these settings, REG3A accumulates at sites of liver injury, localizing specifically to fibrin deposits formed through fibrinogen polymerization, a process triggered by hepatocyte cytolysis. Through its potent free radical scavenging activity, REG3A plays a pivotal role in mitigating local oxidative stress, thereby supporting hepatocyte survival and promoting liver tissue repair and regeneration. [21,22,88].
Under inflammatory conditions where hepatocyte death is prominent, REG3A exerts a potent cytoprotective effect [89]. In vivo, this protection is recapitulated in REG3A transgenic mice and engineered to express human REG3A under the liver-specific albumin promoter. These mice show improved survival in models of acute liver failure induced by acetaminophen or Fas receptor activation [90,91]. These results led to phase 1 and 2 trials of recombinant REG3A for the treatment of acute liver failure [88].
Beyond the liver, REG3A exhibits broad cytoprotective activity across various tissues. In the central nervous system, REG3A safeguards cerebellar, cortical, and hippocampal neurons from H2O2-induced oxidative damage, enhances neuronal plasticity [92,93], and reduces NMDA receptor-mediated excitotoxicity caused by ibotenate [92]. Moreover, REG3A acts as a chemoattractant, recruiting macrophages to sites of neuronal injury, thus supporting repair and regeneration [94]. Similar regenerative effects are observed in cardiac tissue post-myocardial infarction. There, REG3A expression is induced by oncostatin M, an IL-6 family cytokine that drives cardiomyocyte dedifferentiation and remodeling. REG3A promotes macrophage recruitment to protect the cardiac ECM from neutrophil-mediated degradation within the infarcted region, facilitating tissue repair [95,96].
Notably, REG3A also possesses anti-fibrotic properties in both liver and lung tissues. In mouse models, REG3A reduces fibrosis following CCl4-induced liver injury or bile duct ligation [58], as well as bleomycin-induced lung fibrosis [65]. REG3A attenuates oxidative stress and modulates stellate cell responses to TGF-β, thereby preventing excessive epithelial–mesenchymal transition and limiting fibrotic progression [58,65].

5. REG3A in Tissue Regeneration: A Critical Link Between Inflammation and Repair

As previously discussed, the localized recruitment and accumulation of REG3A at sites of tissue injury enables a targeted reduction in oxidative stress and supports tissue repair. The antioxidant function of REG3A underpins its potent pro-survival activity, playing a central role in tissue regeneration across various tissues. In the nervous system, REG3A promotes motor neuron regeneration and exerts neurotrophic effects by enhancing motor neuron survival [93]. It also stimulates Schwann cell proliferation, a crucial step in peripheral nerve repair [61]. Mechanistically, REG3A activates the Akt signaling pathway, contributing to the downstream effects of ciliary neurotrophic factor (CNTF), which is essential for neuronal survival [93]. In liver tissue, REG3A enhances epidermal growth factor (EGF)-induced hepatocyte proliferation, facilitating liver regeneration following partial hepatectomy [89,90,91]. This ability to support healing makes it a promising treatment for liver injury and recovery after surgery. In the skin, REG3A plays a dual role in regeneration and immune modulation. Lai et al. demonstrated that REG3A is expressed in keratinocytes of both the epidermis and hair follicles in psoriasis patients and mouse models. It promotes keratinocyte proliferation while inhibiting terminal differentiation, processes essential for wound re-epithelialization and the hyperproliferative phenotype observed in psoriasis. This mitogenic effect is mediated via the EXTL3-Akt-STAT3 signaling axis [28]. Moreover, REG3A contributes to immune homeostasis in the skin by inducing SHP-1, a negative regulator of Toll-like receptor 3 (TLR-3), thereby suppressing excessive inflammatory responses following skin injury. This regulation is particularly important in diabetic skin inflammation, where hyperglycemia impairs IL-33-dependent REG3A expression, leading a delayed wound healing [43].
Exostosin-like glycosyltransferase 3 (EXTL3), primarily known for regulating heparan sulfate biosynthesis [97], has been proposed as a putative membrane receptor for REG3A. Initially characterized as a receptor for REG1A [98], subsequent bioinformatics analyses and functional studies suggest that EXTL3 also mediates REG3A activity [99]. Binding to EXTL3 activates the RAS-RAF-MEK-ERK signaling cascade in tissues such as the pancreas and skin, supporting cell survival and proliferation [79]. Other receptors have also been implicated in REG3A signaling. These include receptors containing the gp130 subunit and the EGFR, both of which can activate the JAK2/STAT3 pathway, further highlighting the pleiotropic regenerative and immunomodulatory roles of REG3A [75,76].

6. Systemic Roles of REG3A: Linking Metabolism to Health and Disease

Since the discovery of INGAP, the hamster homolog of REG3A, interest has grown in the role of REG family proteins in glucose homeostasis and metabolic regulation. REG3A, in particular, has emerged as a critical factor in pancreatic regeneration and β-cell protection, positioning it as a potential therapeutic target in diabetes and metabolic disorders. Evidence from animal models supports this role. REG3A promotes β-cell survival and may contribute to islet neogenesis via mechanisms involving PI3K/ATF-2/Cyclin D1 signaling, β-cell proliferation, or progenitor cell differentiation [100,101,102,103,104]. Notably, administration of INGAPpp, a pentadecapeptide derived from hamster REG3A, has been shown to increase islet volume and cell mass in diabetic models, preventing streptozotocin (STZ)-induced diabetes without directly stimulating cell proliferation [99,105]. These findings prompted clinical trials in 2009, in which INGAP treatment increased C-peptide levels in type 1 diabetes and lowered HbA1c in type 2 diabetes. However, it did not significantly reduce blood glucose or insulin dependency [106]. Further studies by Xiong et al. confirmed that REG3A does not act as a mitogen for β-cells. In mice expressing REG3A under the RIP-1 promoter, pancreatic islets displayed normal histology and insulin secretion, yet were resistant to STZ-induced hyperglycemia and weight loss [107]. These results reinforce REG3A’s cytoprotective, rather than proliferative, role in β-cell maintenance.
In alcohol-induced metabolic dysfunction, REG3A plays a protective role in maintaining gut–liver axis integrity. Chronic alcohol intake downregulates murine REG3A homologs (Reg3β and Reg3γ), disrupting the spatial segregation between commensal bacteria and the mucosal surface. This leads to bacterial translocation, liver inflammation, and the progression from steatosis to steatohepatitis [59]. REG3A overexpression restores microbial balance and barrier function, attenuating liver injury [59,85]. Similarly, REG3A expression can be induced by prebiotics (e.g., fructo-oligosaccharides), microbial metabolites (e.g., indole-3-acetic acid), or IL-22-producing Lactobacillus reuteri, all of which protect against alcohol-related liver disease [44,59].
Beyond alcohol-related pathology, REG3A also shapes the gut microbiota in obesity and metabolic syndrome. REG3A promotes a Lactobacillus-rich intestinal environment, including strains like Lactobacillus NK318.1, which enhance anti-inflammatory macrophage populations in the gut. These macrophages migrate to adipose tissue, contributing to improved resistance to diet-induced obesity [108]. In a study by Shin et al., a single injection of REG3A preserved gut barrier function, maintained energy balance, and improved glucose regulation in mice on a high-fat diet, suggesting a potential endocrine-like role for REG3A in systemic glucose tolerance [33].
Intriguingly, both alcohol and high-fat diets suppress IL-22 and REG3A expression in the intestine [109]. Yet, intestinal inactivation of murine REG3A homologs does not always correlate with worsened liver outcomes; while bacterial translocation and steatohepatitis were unaffected in one study, endotoxemia was significantly increased following REG3A silencing [110], underscoring a complex and variable role in gut–liver homeostasis.
Finally, REG3A directly contributes to metabolic regulation beyond the gut and pancreas. In liver-specific REG3A transgenic mice, both lean and obese animals showed improved glycemic control and enhanced insulin sensitivity [23]. Similarly, recombinant REG3A administration in obese (ob/ob) or high-fat-diet-fed mice enhanced glucose uptake in skeletal muscle and activated AMPK in a GP130-dependent manner, while reducing protein oxidative damage [23]. These systemic effects support the potential of REG3A as a metabolic modulator affecting multiple organs.

7. REG3A and Cancer: A Dual-Edged Role

REG3A is a critical mediator of cellular survival and growth, with well-established roles in regulating proliferation, differentiation, immune responses, metabolism, and tissue regeneration. These functions are essential for preserving tissue integrity and homeostasis and have consequently drawn attention to REG3A’s potential involvement in cancer biology. Elevated REG3A expression has been reported in certain human tumor types and has been associated with cancer initiation and progression in various experimental models [27,53,57,70,72,74,111,112]. Nevertheless, its exact contribution to tumorigenesis remains to be elucidated, as the literature presents conflicting evidence regarding its pro- and anti-carcinogenic effects, with no clear understanding of phenotypic variations (Table 2).
Members of the REG3 family, including REG3A, have been extensively studied in inflammatory disorders. Their expression is frequently dysregulated in conditions such as pancreatitis, colitis, hepatitis, and cirrhosis, where REG3A appears to function as part of a feedback mechanism aimed at dampening inflammation. Notably, similar patterns of dysregulation are observed in inflammation-associated cancers, such as pancreatic ductal adenocarcinoma, gastrointestinal malignancies, and primary liver cancer, raising the possibility that REG3A could serve as a diagnostic or prognostic biomarker. However, it remains difficult to determine whether REG3A actively promotes cancer or simply responds to changes in inflammation. This is further complicated by the fact that tumors can vary greatly over time and in different areas, and their genetic makeup can change as they grow. These factors underscore the need for large, well-characterized molecular cohorts to accurately assess REG3 protein expression patterns, a requirement that has not yet been fully met in current research.
Emerging evidence suggests that REG3 proteins may be co-opted by both malignant and stromal cells within the tumor microenvironment, where they could modulate antitumor immunity and oncogenic signaling. This potential functional reprogramming highlights the variable role of REG3A in cancer, influenced by factors such as tissue origin, cancer subtype and molecular profile, the pathological status of surrounding tissue, tumor stage, and the immune landscape.
Furthermore, some studies report a negative correlation between REG3A expression and tumor progression in specific settings [20,25,71,113], suggesting that REG3A may act as a tumor suppressor under certain conditions while promoting tumor growth in others. These discrepancies emphasize the highly variable and adaptable role of REG3A in cancer, shaped by the inflammatory milieu and surrounding signaling networks. Table 2 provides a comprehensive overview of the multifaceted role of REG3A in cancer, including a comparative analysis of key findings across various cancer types and experimental models, such as in vitro, in vivo, and human cohort studies.
Several limitations in the current literature hinder a clear understanding of its role in cancer. Most studies rely heavily on in vitro models using diverse cell lines, which may lack biological relevance for a secreted protein like REG3A that operates across multiple cell types and complex systems such as the gut microbiota and metabolism. This may contribute to experimental variability, poor reproducibility, and often contradictory findings. In vivo studies on REG3A remain limited, resulting in significant gaps in our understanding of its functional roles in mammalian cancer models. Notably, conclusions about its tumor-promoting or tumor-suppressive effects often rely on single studies, especially in colorectal, gastric, and liver cancers. To date, pancreatic cancer has received the most attention, with, to our knowledge, approximately ten in vivo studies conducted on three distinct cancer models (Table 2). A further challenge is distinguishing REG3A’s intracellular effects from its extracellular, autocrine, or paracrine functions. This mechanistic uncertainty complicates the interpretation of its biological activity and its role in tumor progression and microenvironmental interactions. Collectively, these limitations present a substantial obstacle to fully elucidating the relevance of REG3A in cancer and underscore the need for replication studies, physiologically relevant models, and integrative research approaches.

7.1. REG3A in Pancreatic Cancer: A Paracrine Oncogenic Role

REG3A is highly expressed in PDAC and is strongly associated with metastatic invasion, poor surgical outcomes, lower progression-free survival, and reduced overall life expectancy (Table 2) [48,77,114,115]. Its expression in pancreatic tissue is significantly elevated in PDAC (up to 24-fold compared to other pancreatic diseases, and 16-fold relative to chronic pancreatitis) [114]. Elevated serum REG3A levels are also detected in PDAC patients and serve as a diagnostic biomarker, distinguishing cases from healthy individuals with 90% sensitivity and 82.8% specificity at cutoff of 18 µg/L [115]. Validation in a cohort of 118 patients further confirmed the association of increased REG3A (both in serum and pancreatic juice) with poor prognosis [114].
Mechanistically, transcriptomic analyses of PDAC cell lines have shown that REG3A upregulates genes involved in cell proliferation and survival programs [116]. Notably, REG3A is primarily secreted by peritumoral pancreatic acinar cells rather than by tumor cells themselves [48,77,114,117]. Acting as a paracrine factor, it activates JAK/STAT and EXTL3/RAS/ERK signaling pathways in both inflamed acinar cells and malignant epithelial cells. In acinar cells, this promotes acinar-to-ductal metaplasia and the formation of pancreatic intraepithelial neoplasia (PanIN) lesions [76], while in cancer cells it enhances tumor growth and perineural invasion [61,70,98,99] (Figure 3). The oncogenic activity of REG3A is counterbalanced by SOCS3, a negative regulator of cytokine signaling. Silencing SOCS3 enhances REG3A-driven tumor progression, while its overexpression suppresses malignancy across multiple pancreatic cancer cell lines [112].

7.2. REG3A in Liver Cancer: A Tumor Suppressive Role Targeting the O-GlcNAc Glycosylation Pathway

REG3A was initially identified in hepatocellular carcinoma (HCC) in 1992 [3], but its role in liver carcinogenesis remained unclear until recently [20]. A number of studies have reported marked overexpression of REG3A in tumor tissue compared to adjacent non-tumor liver, validated at both mRNA and protein levels [27,74]. Elevated REG3A correlates with aggressive features such as larger tumor size and vascular invasion [118], and is found in 24–79% of HCC cases, depending on the detection method [3,27]. Elevated serum REG3A is also observed in 45% of cirrhotic patients and up to 75% with HCC [27]. REG3A expression is enriched in an HCC molecular subtype marked by Wnt/β-catenin activation and CTNNB1 mutations, often linked to early-stage tumors and better prognosis [60,73].
In vitro, REG3A enhances HCC cell proliferation, migration, and invasion via MAPK/ERK and PDGF-ββ signaling [74,118]. However, in vivo studies show a tumor-suppressive function. Moniaux et al. found that hepatic overexpression of REG3A significantly reduces tumor burden and extends survival in both chemical and genetic mouse models [20]. Mechanistically, REG3A reduces O-GlcNAcylation, including MYC modification, by reducing intracellular UDP-GlcNAc levels, thereby promoting MYC phosphorylation and degradation. This effect is independent of OGT, OGA, and GFAT expression or activity, and instead depends on the ability of REG3A to bind hexosamine pathway intermediates via its EPN lectin motif. A mutant (EPN→GPG) form fails to bind these metabolites or reduce O-GlcNAcylation, confirming the importance of carbohydrate-binding. Supporting this, mouse livers expressing wild-type REG3A and non-tumor tissues from HCC patients show reduced UDP-GlcNAc and global O-GlcNAcylation. Clinically, cirrhotic patients with high hepatic REG3A have longer cancer-free survival, although REG3A levels do not affect outcomes once HCC develops [20]. These results suggest that REG3A promotes tumor growth in simple in vitro systems but suppresses tumor formation in vivo by regulating O-GlcNAcylation, a process shaped by systemic metabolism and tissue interactions (Figure 3). These results emphasize the need for physiologically relevant in vivo models that accurately represent human tumors. REG3A exerts pleiotropic functions that extend beyond the tumor itself, influencing organ-level physiology, host–microbiota interactions, and metabolism, factors poorly represented in in vitro systems. Without models that incorporate native tissue architecture and immune components, interpretations of REG3A’s oncogenic potential remain incomplete, limiting progress in therapeutic translation.

7.3. The REG3A Paradox: Conflicting Data Across Solid Tumor Models

The expression of REG3A is dysregulated across multiple human solid tumors, yet its functional role in these cancers remains highly nuanced and varies depending on tumor type and biological conditions (Table 2). In cholangiocarcinoma, REG3A is markedly overexpressed in tumor tissues relative to adjacent non-neoplastic bile duct epithelium, suggesting a possible role in tumor initiation or maintenance [27]. Similarly, in bladder cancer, elevated urinary levels of REG3A have been reported in patients with advanced disease stages. These levels are not only associated with disease progression but also demonstrate promising diagnostic accuracy (sensitivity: 80.2%; specificity: 78.2%) [51]. In contrast, the prognostic and functional relevance of REG3A in lung and head and neck cancers appears more variable. In lung adenocarcinoma, REG3A expression shows no clear correlation with clinical parameters [119]. However, in hypopharyngeal squamous cell carcinoma, high REG3A expression is linked to prolonged patient survival and enhanced sensitivity to chemotherapeutic agents. Supporting these clinical observations, in vitro studies have demonstrated that REG3A exerts anti-proliferative effects and enhances cellular responses to both chemotherapy and radiotherapy [66,120]. The most compelling evidence for the dualistic nature of REG3A comes from studies in gastric and colorectal cancers, where its role oscillates between tumor-suppressive and oncogenic depending on experimental conditions and disease characteristics.
In gastric cancer (GC), several studies report a significant downregulation of REG3A in tumor tissues, implying a tumor-suppressive function [25,71,121]. Functional analyses further reveal that REG3A overexpression inhibits proliferation and induces apoptosis in gastric cancer cell lines, largely through modulation of the PI3K/Akt signaling pathway and upregulation of DMBT1, a known tumor suppressor [71,121]. Conversely, other studies describe elevated REG3A expression in GC tissues, where its knockdown results in decreased cancer cell proliferation, migration, and invasion, indicating a potential oncogenic role under certain conditions [72]. Of note, the sample sizes in the human cohort studies differed significantly: Chen et al. [72] analyzed 41 paired non-tumor/tumor gastric tissue samples, whereas Qiu et al. [71] conducted a large-scale analysis using TCGA, GSE13911, and GSE13861 datasets, encompassing a total of 876 patients with gastric cancer. To date, no further studies have been reported that could clarify the role of REG3A in gastric cancer.
Similar contradictions are observed in colorectal cancer (CRC). One large-scale analysis of 331 matched tumor–normal tissue pairs found REG3A to be enriched in tumors lacking deep invasion, with high expression correlating with improved overall survival [57]. These findings align with data from murine models of colitis-associated cancer, where the murine REG3A homolog suppresses cytokine-driven STAT3 activation, thereby limiting early neoplastic transformation and tumor progression [69]. However, other studies show that REG3A is overexpressed in advanced CRC and is linked to poorer clinical outcomes [70]. Both studies relied on large-scale cohorts, yet they reported opposing correlations between REG3A expression and cancer progression, one showing a negative correlation [57] and the other a positive association [70]. A key methodological difference lies in the choice of control groups: Zheng et al. used pairwise non-tumor tissue adjacent to the tumor [35], whereas Ye et al. compared tumor samples to colonic tissues from a limited number of healthy donors [70]. The imbalance in sample sizes between control and tumor groups (e.g., 6 healthy vs. 90 tumor tissues in GSE33113; 28 healthy vs. 245 tumor tissues in TCGA) may also contribute to the statistical differences found. Similarly, methodological differences in in vivo mouse models are notable. Liu et al. reported a tumor-suppressive role for REG3A, observing a reduced polyp burden in the AOM-DSS-induced colorectal cancer model [69]. In contrast, Ye et al. described a pro-tumoral effect of REG3A following ectopic transplantation of human cancer cells into the flanks of immunodeficient (nude) mice [70]. Mechanistically, using in vitro experiments on the same cancer cell lines, Ye et al. demonstrated that REG3A promotes cancer cell proliferation, migration, and invasion through activation of the AKT and ERK1/2 signaling pathways [70].
At the epigenetic and transcriptional levels, non-coding RNA regulation adds further layers of control. Notably, Yari et al. identified a long non-coding RNA (lncRNA REG1CP) transcribed from a pseudogene located between REG1A and REG1B on chromosome 2p12. REG1CP was shown to interact directly with the REG3A promoter via a triplex RNA/DNA structure, recruiting the helicase FANCJ and facilitating transcriptional activation under the control of the glucocorticoid receptor α [122]. REG1CP is notably upregulated in both preneoplastic colonic lesions and colorectal carcinomas, suggesting a cooperative axis between REG1CP and REG3A in promoting colorectal tumorigenesis.
Collectively, these findings highlight the nuanced and highly variable nature of REG3A’s involvement in cancer. Its function varies not only between tumor types but also within the same malignancy, depending on molecular subtype, tumor stage, and microenvironmental cues. While correlations between REG3A expression and clinical outcomes have been documented across multiple cancer types, mechanistic understanding remains incomplete. Elucidating the signaling networks and regulatory elements that govern REG3A expression and activity will be essential to determine its viability as a diagnostic biomarker, prognostic indicator, or therapeutic target. Given these complexities, future studies should prioritize the use of physiologically relevant in vivo models that preserve tissue-specific architecture, immune interactions, and systemic regulation. Only through such integrative approaches can the full spectrum of REG3A’s oncogenic and tumor-suppressive functions be defined, paving the way for informed translational applications in oncology.

7.4. The REG3A–Gut Microbiota–Inflammation–Cancer Axis: An Emerging Interface in Tumor Biology

Over the past two decades, mounting evidence has highlighted the critical role of the microbiota in cancer development, progression, and therapeutic response (reviewed in [123]). Once viewed as passive bystanders, microbial communities are now understood to be active modulators of host physiology, interacting with genetic, immune, metabolic, and environmental factors to influence carcinogenesis. Among these, the gut microbiome has been most extensively studied, particularly in colorectal cancer. Specific bacterial species, such as Fusobacterium nucleatum, colibactin-producing Escherichia coli, and enterotoxigenic Bacteroides fragilis, exhibit pro-tumorigenic properties [124,125]. These microbes can induce DNA damage, promote chronic inflammation, and reshape immune responses in ways that favor tumor initiation and growth. For example, F. nucleatum adheres to epithelial cells, activates oncogenic β-catenin signaling, and impairs antitumor immunity by recruiting myeloid-derived suppressor cells (MDSCs) and inhibiting natural killer (NK) cell activity [126].
Dysbiosis-driven chronic inflammation is a well-recognized driver of carcinogenesis. Microbial metabolites and ligands for pattern recognition receptors (e.g., Toll-like receptors) can amplify inflammatory responses and disrupt epithelial barriers. In inflammatory bowel disease, an established risk factor for CRC, microbial imbalances closely correlate with disease activity and barrier dysfunction, creating a tumor-permissive microenvironment.
Although direct evidence linking REG3A-modulated microbiota to cancer is currently lacking, REG3A’s known ability to shape a beneficial microbial community and preserve epithelial integrity suggests a potential protective role, particularly in colorectal and liver cancers [86]. REG3A promotes the maintenance of Firmicutes such as Lachnospiraceae and Ruminococcaceae, Gram-positive anaerobes that produce short-chain fatty acids (SCFAs) like butyrate [86]. These metabolites are generally considered protective, they reinforce epithelial barrier function, reduce inflammation, and may help suppress tumorigenesis [127].
Microbial influence on carcinogenesis extends beyond the gut. A well-established example is Helicobacter pylori, a major risk factor for gastric cancer, which contributes to tumorigenesis through chronic inflammation, epigenetic reprogramming, and suppression of tumor suppressor pathways, including those involving DMBT1 and E-cadherin. Notably, REG3A has been identified as a positive regulator of DMBT1, potentially counteracting H. pylori-mediated silencing of this tumor suppressor [128]. Whether this effect results from direct transcriptional regulation or is secondary to REG3A’s antimicrobial activity remains unresolved. The observed upregulation of REG3A in H. pylori-infected gastric tissue suggests a potential protective role, but further research is needed to clarify its function [129].
In summary, the REG3A-microbiota–cancer axis represents a promising but largely unexplored domain. While its impact on tumorigenesis remains to be definitively established, existing evidence suggests that REG3A may influence cancer risk by modulating microbial composition, inflammatory signaling, and epithelial integrity. Bridging this knowledge gap presents a valuable opportunity for future investigation.

8. Clinical Relevance and Therapeutic Perspectives

Despite a robust body of preclinical evidence supporting the involvement of REG3A in diverse pathological settings, including inflammation, tissue regeneration and metabolic disorders, its translation to clinical application remains nascent. To date, only a few clinical trials have investigated REG3A or related peptides in humans. Nevertheless, early-phase studies offer encouraging translational insights and suggest the potential for broader therapeutic utility. One such example involves INGAP peptide (INGAPpp), a structural homolog of REG3A derived from hamster, which has been evaluated in two randomized, placebo-controlled Phase II clinical trials targeting diabetes mellitus. In type 1 diabetes (T1D) (NCT00071409), 63 patients received daily subcutaneous injections of 300 mg or 600 mg INGAP for 90 days [106]. The primary outcome, arginine-stimulated C-peptide via mixed-meal tolerance test, showed a significant increase in the 600 mg group compared to placebo (p = 0.0058), indicating partial β-cell functional recovery. Secondary outcomes, including HbA1c and insulin dose, improved modestly. Approximately 50% of the treatment effect persisted after a 30-day washout, suggesting transient β-cell preservation. Injection-site reactions were common (up to 90%), but no serious systemic adverse events or immunogenic responses were reported. A parallel Phase II study in type 2 diabetes (T2D), involving 126 participants, confirmed these findings [106]. The 600 mg group demonstrated significantly elevated C-peptide levels (p = 0.031) and a 0.9% reduction in HbA1c at day 90 (vs. 0.47% in placebo, p = 0.009), with durable metabolic effects persisting at follow-up (p = 0.013). Side effects were again limited primarily to injection-site reactions (about 65%), without notable systemic toxicity.
A second approach has explored recombinant human REG3A protein (ALF 5755) in the treatment of acute liver injury. A Phase IIa randomized, double-blind, placebo-controlled trial (NCT01318525) enrolled 57 adults with severe acute hepatitis (SAH) or early-stage acute liver failure (ALF) of non-acetaminophen etiology [88]. ALF 5755 (5 mg/kg, IV) was evaluated for its impact on coagulation recovery (change in prothrombin ratio over 72 h). While overall results across all etiologies were neutral, a subgroup analysis in HBV and autoimmune hepatitis patients revealed a significantly improved prothrombin slope (p = 0.04) and a reduction in hospitalization duration (8 vs. 14 days, p = 0.02). Prior Phase I data confirmed that ALF 5755 was well-tolerated in healthy volunteers, with no serious adverse events, acceptable pharmacokinetics, and no immunogenicity [21].
Together, these studies provide first-in-human validation of the regenerative, anti-inflammatory and metabolic potential of REG3A-derived therapeutics. While clinical indications to date have focused on diabetes and ALF, REG3A’s broader biological properties, including epithelial repair, preservation of intestinal barrier integrity, and modulation of innate immunity and the gut microbiota, point to its therapeutic promise in other conditions, such as mucosal injury and chronic inflammatory diseases. Its potential relevance to oncology is particularly compelling, given REG3A’s roles in immune modulation, epithelial regeneration, and microenvironmental remodeling. However, due to its variable effects, such as evidence of pro-tumorigenic activity in certain malignancies, careful consideration of disease type, underlying mechanisms and target cell populations is essential. Rigorous preclinical and translational studies are needed to delineate its safety profile and therapeutic window before advancing REG3A-based interventions in cancer. Moreover, recent advances in cancer immunotherapy, alongside growing evidence that gut microbiota composition influences treatment efficacy, highlight a promising opportunity for REG3A-based microbiota modulation to enhance immunotherapeutic outcomes. By reinforcing epithelial integrity and restoring a favorable microbial milieu, REG3A could synergize with immune checkpoint inhibitors to strengthen antitumor immunity and improve patient response rates.
In summary, REG3A-based interventions are emerging as promising tools in regenerative medicine, metabolic disorders, and potentially oncology, supported by early clinical safety and efficacy signals. However, conflicting data and concerns about REG3A’s oncogenic potential have slowed progress in cancer applications. Future studies should focus on patient selection, dose optimization, long-term safety, and understanding tissue-specific effects to fully unlock the clinical benefits of REG3A across diseases.

9. Conclusions

REG3A is a multifaceted molecule with critical and diverse roles in both health and disease. Physiologically, it plays a central role in the gastrointestinal tract by supporting antimicrobial defense, promoting epithelial repair, and maintaining mucosal homeostasis. In contrast, its involvement in cancer is complex and highly variable, with REG3A frequently dysregulated and linked to chronic inflammation, immune evasion, and aberrant cell proliferation, factors that complicate efforts to define a universal role in tumorigenesis. Despite gaps in our understanding of its biology, the diverse functions of REG3A highlight its strong clinical relevance. Its immunomodulatory and regenerative properties position it as a promising target for treating inflammatory diseases characterized by epithelial damage and remodeling. In oncology, its dual role as either a tumor promoter or suppressor depends on tumor type, cellular features, and signals from the surrounding microenvironment. Mechanistically, REG3A influences key oncogenic signaling pathways and metabolic reprogramming, notably glucose metabolism essential for cancer cell survival and proliferation. Furthermore, by modulating host–microbiota interactions, REG3A contributes to shaping tumor immunity and systemic inflammation, broadening its relevance in cancer biology and targeted therapy.
Importantly, REG3A has completed Phase 1 and 2 clinical trials for acute liver failure and diabetes, demonstrating favorable safety profiles. Nonetheless, its development as a therapeutic agent requires rigorous safety evaluation, especially for systemic or long-term use. Unlocking the full clinical potential of REG3A will require integrative research approaches that combine mechanistic insight with appropriate translational models to guide safe and effective applications. This will pave the way for innovative therapies harnessing the unique biology of REG3A in both regenerative medicine and precision oncology.

Author Contributions

Conceptualization, J.F. and N.M.; writing—original draft preparation, J.F. and N.M.; writing—review and editing, J.F., H.S., T.S.N., H.X. and N.M.; funding acquisition, J.F. and N.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review received no external funding.

Acknowledgments

We are deeply grateful to the many colleagues and collaborators, past and present, whose contributions have enriched our research on REG3A. Their insight and dedication have been invaluable. We extend special thanks to Christian Bréchot for his continued support and advice. We also sincerely thank our public and private funders, especially the Agence Nationale de la Recherche (ANR), ARC Foundation for Cancer Research, Ligue Nationale Contre le Cancer, Gefluc (Entreprises contre le cancer), INSERM, the Doctoral School of Life and Health Sciences at Paris-Saclay University, and ALFACT Innovation, whose support was essential to the completion of this research.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AKTProtein kinase B
ALFAcute liver failure
AMPAntimicrobial protein
AMPKAdenosine monophosphate-activated protein kinase
CCL4Tetrachloromethane
CRCColorectal cancer
CRDCarbohydrate-recognition domain
DAMPsDamage-associated molecular patterns
DMBT1Deleted in malignant brain tumors 1
ECMExtracellular matrix
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
ERKExtracellular signal-regulated kinase
EXTL3Exostosin-like glycosyltransferase 3
FOXM1Forkhead box M1
GCGastric cancer
GFATGlucosamine:fructose-6-phosphate aminotransferase
GlcNAcN-acetylglucosamine
HCCHepatocellular carcinoma
HIPHepatocarcinoma-intestine-pancreas associated protein
INGAPIslet neogenesis-associated protein
JAKJanus kinase
LncRNALong non-coding RNA
LPSLipopolysaccharide
MAPKMitogen-activated protein kinase
NMDAN-methyl-D-aspartic acid
OGAO-GlcNAcase
OGTO-GlcNAc transferase
PAMPsPathogen-associated molecular patterns
PAPPancreatitis-associated protein
PDACPancreatic ductal adenocarcinoma
PDBProtein data bank
PDGFPlatelet-derived growth factor
PTPPancreatic thread protein
REGRegenerating gene
ROSReactive oxygen species
SHP-1Src homology region 2 (SH2) domain-containing phosphatase-1
SOCS3Suppressor of cytokine signaling 3
STAT-3Signal transducer and activator of transcription-3
TNFαTumor necrosis factor α

References

  1. Keim, V.; Rohr, G.; Stöckert, H.G.; Haberich, F.J. An Additional Secretory Protein in the Rat Pancreas. Digestion 1984, 29, 242–249. [Google Scholar] [CrossRef] [PubMed]
  2. Keim, V.; Iovanna, J.L.; Rohr, G.; Usadel, K.H.; Dagorn, J.C. Characterization of a Rat Pancreatic Secretory Protein Associated with Pancreatitis. Gastroenterology 1991, 100, 775–782. [Google Scholar] [CrossRef] [PubMed]
  3. Lasserre, C.; Christa, L.; Simon, M.T.; Vernier, P.; Bréchot, C. A Novel Gene (HIP) Activated in Human Primary Liver Cancer. Cancer Res. 1992, 52, 5089–5095. [Google Scholar] [PubMed]
  4. Unno, M.; Yonekura, H.; Nakagawara, K.; Watanabe, T.; Miyashita, H.; Moriizumi, S.; Okamoto, H.; Itoh, T.; Teraoka, H. Structure, Chromosomal Localization, and Expression of Mouse Reg Genes, Reg I and Reg II. A Novel Type of Reg Gene, Reg II, Exists in the Mouse Genome. J. Biol. Chem. 1993, 268, 15974–15982. [Google Scholar] [CrossRef]
  5. Nata, K.; Liu, Y.; Xu, L.; Ikeda, T.; Akiyama, T.; Noguchi, N.; Kawaguchi, S.; Yamauchi, A.; Takahashi, I.; Shervani, N.J.; et al. Molecular Cloning, Expression and Chromosomal Localization of a Novel Human REG Family Gene, REG III. Gene 2004, 340, 161–170. [Google Scholar] [CrossRef]
  6. Watanabe, T.; Yonekura, H.; Terazono, K.; Yamamoto, H.; Okamoto, H. Complete Nucleotide Sequence of Human Reg Gene and Its Expression in Normal and Tumoral Tissues. The Reg Protein, Pancreatic Stone Protein, and Pancreatic Thread Protein Are One and the Same Product of the Gene. J. Biol. Chem. 1990, 265, 7432–7439. [Google Scholar] [CrossRef]
  7. Moriizumi, S.; Watanabe, T.; Unno, M.; Nakagawara, K.; Suzuki, Y.; Miyashita, H.; Yonekura, H.; Okamoto, H. Isolation, Structural Determination and Expression of a Novel Reg Gene, Human regI Beta. Biochim. Biophys. Acta 1994, 1217, 199–202. [Google Scholar] [CrossRef]
  8. Lasserre, C.; Simon, M.T.; Ishikawa, H.; Diriong, S.; Nguyen, V.C.; Christa, L.; Vernier, P.; Brechot, C. Structural Organization and Chromosomal Localization of a Human Gene (HIP/PAP) Encoding a C-Type Lectin Overexpressed in Primary Liver Cancer. Eur. J. Biochem. 1994, 224, 29–38. [Google Scholar] [CrossRef]
  9. Iovanna, J.; Orelle, B.; Keim, V.; Dagorn, J.C. Messenger RNA Sequence and Expression of Rat Pancreatitis-Associated Protein, a Lectin-Related Protein Overexpressed during Acute Experimental Pancreatitis. J. Biol. Chem. 1991, 266, 24664–24669. [Google Scholar] [CrossRef]
  10. Abergel, C.; Chenivesse, S.; Stinnakre, M.G.; Guasco, S.; Bréchot, C.; Claverie, J.M.; Devinoy, E.; Christa, L. Crystallization and Preliminary Crystallographic Study of HIP/PAP, a Human C-Lectin Overexpressed in Primary Liver Cancers. Acta Crystallogr. D Biol. Crystallogr. 1999, 55, 1487–1489. [Google Scholar] [CrossRef]
  11. Mukherjee, S.; Partch, C.L.; Lehotzky, R.E.; Whitham, C.V.; Chu, H.; Bevins, C.L.; Gardner, K.H.; Hooper, L.V. Regulation of C-Type Lectin Antimicrobial Activity by a Flexible N-Terminal Prosegment. J. Biol. Chem. 2009, 284, 4881–4888. [Google Scholar] [CrossRef]
  12. Graf, R.; Schiesser, M.; Scheele, G.A.; Marquardt, K.; Frick, T.W.; Ammann, R.W.; Bimmler, D. A Family of 16-kDa Pancreatic Secretory Stress Proteins Form Highly Organized Fibrillar Structures upon Tryptic Activation. J. Biol. Chem. 2001, 276, 21028–21038. [Google Scholar] [CrossRef]
  13. Medveczky, P.; Szmola, R.; Sahin-Tóth, M. Proteolytic Activation of Human Pancreatitis-Associated Protein Is Required for Peptidoglycan Binding and Bacterial Aggregation. Biochem. J. 2009, 420, 335–343. [Google Scholar] [CrossRef]
  14. Schiesser, M.; Bimmler, D.; Frick, T.W.; Graf, R. Conformational Changes of Pancreatitis-Associated Protein (PAP) Activated by Trypsin Lead to Insoluble Protein Aggregates. Pancreas 2001, 22, 186–192. [Google Scholar] [CrossRef]
  15. Ho, M.-R.; Lou, Y.-C.; Lin, W.-C.; Lyu, P.-C.; Huang, W.-N.; Chen, C. Human Pancreatitis-Associated Protein Forms Fibrillar Aggregates with a Native-like Conformation. J. Biol. Chem. 2006, 281, 33566–33576. [Google Scholar] [CrossRef] [PubMed]
  16. Christa, L.; Felin, M.; Morali, O.; Simon, M.T.; Lasserre, C.; Brechot, C.; Sève, A.P. The Human HIP Gene, Overexpressed in Primary Liver Cancer Encodes for a C-Type Carbohydrate Binding Protein with Lactose Binding Activity. FEBS Lett. 1994, 337, 114–118. [Google Scholar] [CrossRef] [PubMed]
  17. Drickamer, K. C-Type Lectin-like Domains. Curr. Opin. Struct. Biol. 1999, 9, 585–590. [Google Scholar] [CrossRef] [PubMed]
  18. Lehotzky, R.E.; Partch, C.L.; Mukherjee, S.; Cash, H.L.; Goldman, W.E.; Gardner, K.H.; Hooper, L.V. Molecular Basis for Peptidoglycan Recognition by a Bactericidal Lectin. Proc. Natl. Acad. Sci. USA 2010, 107, 7722–7727. [Google Scholar] [CrossRef]
  19. Cash, H.L.; Whitham, C.V.; Behrendt, C.L.; Hooper, L.V. Symbiotic Bacteria Direct Expression of an Intestinal Bactericidal Lectin. Science 2006, 313, 1126–1130. [Google Scholar] [CrossRef]
  20. Moniaux, N.; Geoffre, N.; Deshayes, A.; Dos Santos, A.; Job, S.; Lacoste, C.; Nguyen, T.-S.; Darnaud, M.; Friedel-Arboleas, M.; Guettier, C.; et al. Tumor Suppressive Role of the Antimicrobial Lectin REG3A Targeting the O -GlcNAc Glycosylation Pathway. Hepatology 2025, 81, 1416–1432. [Google Scholar] [CrossRef]
  21. Moniaux, N.; Song, H.; Darnaud, M.; Garbin, K.; Gigou, M.; Mitchell, C.; Samuel, D.; Jamot, L.; Amouyal, P.; Amouyal, G.; et al. Human Hepatocarcinoma-Intestine-Pancreas/Pancreatitis-Associated Protein Cures Fas-Induced Acute Liver Failure in Mice by Attenuating Free-Radical Damage in Injured Livers. Hepatology 2011, 53, 618–627. [Google Scholar] [CrossRef]
  22. Moniaux, N.; Darnaud, M.; Garbin, K.; Dos Santos, A.; Guettier, C.; Samuel, D.; Amouyal, G.; Amouyal, P.; Bréchot, C.; Faivre, J. The Reg3α (HIP/PAP) Lectin Suppresses Extracellular Oxidative Stress in a Murine Model of Acute Liver Failure. PLoS ONE 2015, 10, e0125584. [Google Scholar] [CrossRef]
  23. Gonzalez, P.; Dos Santos, A.; Darnaud, M.; Moniaux, N.; Rapoud, D.; Lacoste, C.; Nguyen, T.-S.; Moullé, V.S.; Deshayes, A.; Amouyal, G.; et al. Antimicrobial Protein REG3A Regulates Glucose Homeostasis and Insulin Resistance in Obese Diabetic Mice. Commun. Biol. 2023, 6, 269. [Google Scholar] [CrossRef]
  24. Hervieu, V.; Christa, L.; Gouysse, G.; Bouvier, R.; Chayvialle, J.-A.; Bréchot, C.; Scoazec, J.-Y. HIP/PAP, a Member of the Reg Family, Is Expressed in Glucagon-Producing Enteropancreatic Endocrine Cells and Tumors. Hum. Pathol. 2006, 37, 1066–1075. [Google Scholar] [CrossRef]
  25. Choi, B.; Suh, Y.; Kim, W.-H.; Christa, L.; Park, J.; Bae, C.-D. Downregulation of Regenerating Islet-Derived 3 Alpha (REG3A) in Primary Human Gastric Adenocarcinomas. Exp. Mol. Med. 2007, 39, 796–804. [Google Scholar] [CrossRef]
  26. Christa, L.; Carnot, F.; Simon, M.T.; Levavasseur, F.; Stinnakre, M.G.; Lasserre, C.; Thepot, D.; Clement, B.; Devinoy, E.; Brechot, C. HIP/PAP Is an Adhesive Protein Expressed in Hepatocarcinoma, Normal Paneth, and Pancreatic Cells. Am. J. Physiol. 1996, 271, G993–G1002. [Google Scholar] [CrossRef] [PubMed]
  27. Christa, L.; Simon, M.T.; Brezault-Bonnet, C.; Bonte, E.; Carnot, F.; Zylberberg, H.; Franco, D.; Capron, F.; Roskams, T.; Bréchot, C. Hepatocarcinoma-Intestine-Pancreas/Pancreatic Associated Protein (HIP/PAP) Is Expressed and Secreted by Proliferating Ductules as Well as by Hepatocarcinoma and Cholangiocarcinoma Cells. Am. J. Pathol. 1999, 155, 1525–1533. [Google Scholar] [CrossRef]
  28. Lai, Y.; Li, D.; Li, C.; Muehleisen, B.; Radek, K.A.; Park, H.J.; Jiang, Z.; Li, Z.; Lei, H.; Quan, Y.; et al. The Antimicrobial Protein REG3A Regulates Keratinocyte Proliferation and Differentiation after Skin Injury. Immunity 2012, 37, 74–84. [Google Scholar] [CrossRef] [PubMed]
  29. Chakraborty, C.; Vrontakis, M.; Molnar, P.; Schroedter, I.C.; Katsumata, N.; Murphy, L.J.; Shiu, R.P.; Friesen, H.G. Expression of Pituitary Peptide 23 in the Rat Uterus: Regulation by Estradiol. Mol. Cell. Endocrinol. 1995, 108, 149–154. [Google Scholar] [CrossRef] [PubMed]
  30. Tachibana, K.; Marquardt, H.; Yokoya, S.; Friesen, H.G. Growth Hormone-Releasing Hormone Stimulates and Somatostatin Inhibits the Release of a Novel Protein by Cultured Rat Pituitary Cells. Mol. Endocrinol. 1988, 2, 973–978. [Google Scholar] [CrossRef]
  31. Brandl, K.; Plitas, G.; Mihu, C.N.; Ubeda, C.; Jia, T.; Fleisher, M.; Schnabl, B.; DeMatteo, R.P.; Pamer, E.G. Vancomycin-Resistant Enterococci Exploit Antibiotic-Induced Innate Immune Deficits. Nature 2008, 455, 804–807. [Google Scholar] [CrossRef]
  32. Natividad, J.M.M.; Hayes, C.L.; Motta, J.-P.; Jury, J.; Galipeau, H.J.; Philip, V.; Garcia-Rodenas, C.L.; Kiyama, H.; Bercik, P.; Verdu, E.F. Differential Induction of Antimicrobial REGIII by the Intestinal Microbiota and Bifidobacterium Breve NCC2950. Appl. Environ. Microbiol. 2013, 79, 7745–7754. [Google Scholar] [CrossRef]
  33. Shin, J.H.; Bozadjieva-Kramer, N.; Shao, Y.; Lyons-Abbott, S.; Rupp, A.C.; Sandoval, D.A.; Seeley, R.J. The Gut Peptide Reg3g Links the Small Intestine Microbiome to the Regulation of Energy Balance, Glucose Levels, and Gut Function. Cell Metab. 2022, 34, 1765–1778.e6. [Google Scholar] [CrossRef]
  34. Vaishnava, S.; Behrendt, C.L.; Ismail, A.S.; Eckmann, L.; Hooper, L.V. Paneth Cells Directly Sense Gut Commensals and Maintain Homeostasis at the Intestinal Host-Microbial Interface. Proc. Natl. Acad. Sci. USA 2008, 105, 20858–20863. [Google Scholar] [CrossRef]
  35. Zheng, Y.; Valdez, P.A.; Danilenko, D.M.; Hu, Y.; Sa, S.M.; Gong, Q.; Abbas, A.R.; Modrusan, Z.; Ghilardi, N.; de Sauvage, F.J.; et al. Interleukin-22 Mediates Early Host Defense against Attaching and Effacing Bacterial Pathogens. Nat. Med. 2008, 14, 282–289. [Google Scholar] [CrossRef]
  36. Vaishnava, S.; Yamamoto, M.; Severson, K.M.; Ruhn, K.A.; Yu, X.; Koren, O.; Ley, R.; Wakeland, E.K.; Hooper, L.V. The Antibacterial Lectin RegIIIgamma Promotes the Spatial Segregation of Microbiota and Host in the Intestine. Science 2011, 334, 255–258. [Google Scholar] [CrossRef]
  37. van Ampting, M.T.J.; Rodenburg, W.; Vink, C.; Kramer, E.; Schonewille, A.J.; Keijer, J.; van der Meer, R.; Bovee-Oudenhoven, I.M.J. Ileal Mucosal and Fecal Pancreatitis Associated Protein Levels Reflect Severity of Salmonella Infection in Rats. Dig. Dis. Sci. 2009, 54, 2588–2597. [Google Scholar] [CrossRef] [PubMed]
  38. van Ampting, M.T.J.; Loonen, L.M.P.; Schonewille, A.J.; Konings, I.; Vink, C.; Iovanna, J.; Chamaillard, M.; Dekker, J.; van der Meer, R.; Wells, J.M.; et al. Intestinally Secreted C-Type Lectin Reg3b Attenuates Salmonellosis but Not Listeriosis in Mice. Infect. Immun. 2012, 80, 1115–1120. [Google Scholar] [CrossRef] [PubMed]
  39. Loonen, L.M.P.; Stolte, E.H.; Jaklofsky, M.T.J.; Meijerink, M.; Dekker, J.; van Baarlen, P.; Wells, J.M. REG3γ-Deficient Mice Have Altered Mucus Distribution and Increased Mucosal Inflammatory Responses to the Microbiota and Enteric Pathogens in the Ileum. Mucosal. Immunol. 2014, 7, 939–947. [Google Scholar] [CrossRef] [PubMed]
  40. Dusetti, N.J.; Ortiz, E.M.; Mallo, G.V.; Dagorn, J.C.; Iovanna, J.L. Pancreatitis-Associated Protein I (PAP I), an Acute Phase Protein Induced by Cytokines. Identification of Two Functional Interleukin-6 Response Elements in the Rat PAP I Promoter Region. J. Biol. Chem. 1995, 270, 22417–22421. [Google Scholar] [CrossRef]
  41. Gurr, W.; Yavari, R.; Wen, L.; Shaw, M.; Mora, C.; Christa, L.; Sherwin, R.S. A Reg Family Protein Is Overexpressed in Islets from a Patient with New-Onset Type 1 Diabetes and Acts as T-Cell Autoantigen in NOD Mice. Diabetes 2002, 51, 339–346. [Google Scholar] [CrossRef]
  42. Yoshino, N.; Ishihara, S.; Rumi, M.A.K.; Ortega-Cava, C.F.; Yuki, T.; Kazumori, H.; Takazawa, S.; Okamoto, H.; Kadowaki, Y.; Kinoshita, Y. Interleukin-8 Regulates Expression of Reg Protein in Helicobacter Pylori-Infected Gastric Mucosa. Am. J. Gastroenterol. 2005, 100, 2157–2166. [Google Scholar] [CrossRef]
  43. Wu, Y.; Quan, Y.; Liu, Y.; Liu, K.; Li, H.; Jiang, Z.; Zhang, T.; Lei, H.; Radek, K.A.; Li, D.; et al. Hyperglycaemia Inhibits REG3A Expression to Exacerbate TLR3-Mediated Skin Inflammation in Diabetes. Nat. Commun. 2016, 7, 13393. [Google Scholar] [CrossRef]
  44. Hendrikx, T.; Duan, Y.; Wang, Y.; Oh, J.-H.; Alexander, L.M.; Huang, W.; Stärkel, P.; Ho, S.B.; Gao, B.; Fiehn, O.; et al. Bacteria Engineered to Produce IL-22 in Intestine Induce Expression of REG3G to Reduce Ethanol-Induced Liver Disease in Mice. Gut 2019, 68, 1504–1515. [Google Scholar] [CrossRef] [PubMed]
  45. Gironella, M.; Iovanna, J.L.; Sans, M.; Gil, F.; Peñalva, M.; Closa, D.; Miquel, R.; Piqué, J.M.; Panés, J. Anti-Inflammatory Effects of Pancreatitis Associated Protein in Inflammatory Bowel Disease. Gut 2005, 54, 1244–1253. [Google Scholar] [CrossRef] [PubMed]
  46. Ferrara, J.L.M.; Harris, A.C.; Greenson, J.K.; Braun, T.M.; Holler, E.; Teshima, T.; Levine, J.E.; Choi, S.W.J.; Huber, E.; Landfried, K.; et al. Regenerating Islet-Derived 3-Alpha Is a Biomarker of Gastrointestinal Graft-versus-Host Disease. Blood 2011, 118, 6702–6708. [Google Scholar] [CrossRef]
  47. Ding, X.; Liu, S.; Zhu, S.; Zhou, B.; Ma, H. A Pilot Study on Human Circulating System Indicated That Regenerating Islet-Derived Protein 3 Gamma (REG3A) Is a Potential Prognostic Biomarker for Sepsis. Am. J. Cardiol. 2023, 190, 90–95. [Google Scholar] [CrossRef]
  48. Nigri, J.; Gironella, M.; Bressy, C.; Vila-Navarro, E.; Roques, J.; Lac, S.; Bontemps, C.; Kozaczyk, C.; Cros, J.; Pietrasz, D.; et al. PAP/REG3A Favors Perineural Invasion in Pancreatic Adenocarcinoma and Serves as a Prognostic Marker. Cell. Mol. Life Sci. 2017, 74, 4231–4243. [Google Scholar] [CrossRef]
  49. DePriest, B.P.; Li, H.; Bidgoli, A.; Onstad, L.; Couriel, D.; Lee, S.J.; Paczesny, S. Regenerating Islet-Derived Protein 3-α Is a Prognostic Biomarker for Gastrointestinal Chronic Graft-versus-Host Disease. Blood Adv. 2022, 6, 2981–2986. [Google Scholar] [CrossRef]
  50. Li, W.-S.; Chen, T.-J.; Lee, S.-W.; Yang, C.-C.; Tian, Y.-F.; Kuo, Y.-H.; Tsai, H.-H.; Wu, L.-C.; Yeh, C.-F.; Shiue, Y.-L.; et al. REG3A Overexpression Functions as a Negative Predictive and Prognostic Biomarker in Rectal Cancer Patients Receiving CCRT. Histol. Histopathol. 2024, 39, 91–104. [Google Scholar] [CrossRef]
  51. Nitta, Y.; Konishi, H.; Makino, T.; Tanaka, T.; Kawashima, H.; Iovanna, J.L.; Nakatani, T.; Kiyama, H. Urinary Levels of Hepatocarcinoma-Intestine-Pancreas/Pancreatitis-Associated Protein as a Diagnostic Biomarker in Patients with Bladder Cancer. BMC Urol. 2012, 12, 24. [Google Scholar] [CrossRef]
  52. Sarles, J.; Barthellemy, S.; Férec, C.; Iovanna, J.; Roussey, M.; Farriaux, J.P.; Toutain, A.; Berthelot, J.; Maurin, N.; Codet, J.P.; et al. Blood Concentrations of Pancreatitis Associated Protein in Neonates: Relevance to Neonatal Screening for Cystic Fibrosis. Arch. Dis. Child. Fetal Neonatal Ed. 1999, 80, F118–F122. [Google Scholar] [CrossRef]
  53. Xie, M.-J.; Motoo, Y.; Iovanna, J.L.; Su, S.-B.; Ohtsubo, K.; Matsubara, F.; Sawabu, N. Overexpression of Pancreatitis-Associated Protein (PAP) in Human Pancreatic Ductal Adenocarcinoma. Dig. Dis. Sci. 2003, 48, 459–464. [Google Scholar] [CrossRef] [PubMed]
  54. Matsumura, N.; Zembutsu, H.; Yamaguchi, K.; Sasaki, K.; Tsuruma, T.; Nishidate, T.; Denno, R.; Hirata, K. Identification of Novel Molecular Markers for Detection of Gastric Cancer Cells in the Peripheral Blood Circulation Using Genome-Wide Microarray Analysis. Exp. Ther. Med. 2011, 2, 705–713. [Google Scholar] [CrossRef] [PubMed]
  55. Ogawa, H.; Fukushima, K.; Naito, H.; Funayama, Y.; Unno, M.; Takahashi, K.; Kitayama, T.; Matsuno, S.; Ohtani, H.; Takasawa, S.; et al. Increased Expression of HIP/PAP and Regenerating Gene III in Human Inflammatory Bowel Disease and a Murine Bacterial Reconstitution Model. Inflamm. Bowel Dis. 2003, 9, 162–170. [Google Scholar] [CrossRef] [PubMed]
  56. Marafini, I.; Di Sabatino, A.; Zorzi, F.; Monteleone, I.; Sedda, S.; Cupi, M.L.; Antenucci, C.; Biancheri, P.; Giuffrida, P.; Di Stefano, M.; et al. Serum Regenerating Islet-Derived 3-Alpha Is a Biomarker of Mucosal Enteropathies. Aliment. Pharmacol. Ther. 2014, 40, 974–981. [Google Scholar] [CrossRef]
  57. Zheng, H.; Sugawara, A.; Okamoto, H.; Takasawa, S.; Takahashi, H.; Masuda, S.; Takano, Y. Expression Profile of the REG Gene Family in Colorectal Carcinoma. J. Histochem. Cytochem. 2011, 59, 106–115. [Google Scholar] [CrossRef]
  58. Li, Q.; Li, H.; Lv, Y.; Zhang, Q.; Zhang, X.; Li, S.; Zheng, X.; Wang, Y.; Hao, Z. Hepatocarcinoma-Intestine-Pancreas/Pancreatitis-Associated Protein (HIP/PAP) Confers Protection against Hepatic Fibrosis through Downregulation of Transforming Growth Factor β Receptor II. Lab. Investig. 2020, 100, 466–482. [Google Scholar] [CrossRef]
  59. Yan, A.W.; Fouts, D.E.; Brandl, J.; Stärkel, P.; Torralba, M.; Schott, E.; Tsukamoto, H.; Nelson, K.E.; Brenner, D.A.; Schnabl, B. Enteric Dysbiosis Associated with a Mouse Model of Alcoholic Liver Disease. Hepatology 2011, 53, 96–105. [Google Scholar] [CrossRef]
  60. Cavard, C.; Terris, B.; Grimber, G.; Christa, L.; Audard, V.; Radenen-Bussiere, B.; Simon, M.-T.; Renard, C.-A.; Buendia, M.-A.; Perret, C. Overexpression of Regenerating Islet-Derived 1 Alpha and 3 Alpha Genes in Human Primary Liver Tumors with Beta-Catenin Mutations. Oncogene 2006, 25, 599–608. [Google Scholar] [CrossRef]
  61. Livesey, F.J.; O’Brien, J.A.; Li, M.; Smith, A.G.; Murphy, L.J.; Hunt, S.P. A Schwann Cell Mitogen Accompanying Regeneration of Motor Neurons. Nature 1997, 390, 614–618. [Google Scholar] [CrossRef] [PubMed]
  62. Ampo, K.-I.; Suzuki, A.; Konishi, H.; Kiyama, H. Induction of Pancreatitis-Associated Protein (PAP) Family Members in Neurons after Traumatic Brain Injury. J. Neurotrauma 2009, 26, 1683–1693. [Google Scholar] [CrossRef] [PubMed]
  63. Duplan, L.; Michel, B.; Boucraut, J.; Barthellémy, S.; Desplat-Jego, S.; Marin, V.; Gambarelli, D.; Bernard, D.; Berthézène, P.; Alescio-Lautier, B.; et al. Lithostathine and Pancreatitis-Associated Protein Are Involved in the Very Early Stages of Alzheimer’s Disease. Neurobiol. Aging 2001, 22, 79–88. [Google Scholar] [CrossRef] [PubMed]
  64. Bartoli, C.; Baeza, N.; Figarella, C.; Pellegrini, I.; Figarella-Branger, D. Expression of Peptide-23/Pancreatitis-Associated Protein and Reg Genes in Human Pituitary and Adenomas: Comparison with Other Fetal and Adult Human Tissues. J. Clin. Endocrinol. Metab. 1998, 83, 4041–4046. [Google Scholar] [CrossRef]
  65. Zheng, X.; Li, Q.; Tian, H.; Li, H.; Lv, Y.; Wang, Y.; He, L.; Huo, Y.; Hao, Z. HIP/PAP Protects against Bleomycin-Induced Lung Injury and Inflammation and Subsequent Fibrosis in Mice. J. Cell. Mol. Med. 2020, 24, 6804–6821. [Google Scholar] [CrossRef]
  66. Masui, T.; Ota, I.; Itaya-Hironaka, A.; Takeda, M.; Kasai, T.; Yamauchi, A.; Sakuramoto-Tsuchida, S.; Mikami, S.; Yane, K.; Takasawa, S.; et al. Expression of REG III and Prognosis in Head and Neck Cancer. Oncol. Rep. 2013, 30, 573–578. [Google Scholar] [CrossRef]
  67. You, G.; Cao, H.; Yan, L.; He, P.; Wang, Y.; Liu, B.; Shao, F. MicroRNA-10a-3p Mediates Th17/Treg Cell Balance and Improves Renal Injury by Inhibiting REG3A in Lupus Nephritis. Int. Immunopharmacol. 2020, 88, 106891. [Google Scholar] [CrossRef]
  68. Jiang, T.; Huang, Y.; Liu, H.; Xu, Q.; Gong, Y.; Chen, Y.; Hu, X.; Han, Z.; Gao, M. Reduced miR-146a Promotes REG3A Expression and Macrophage Migration in Polymyositis and Dermatomyositis. Front. Immunol. 2020, 11, 37. [Google Scholar] [CrossRef]
  69. Liu, X.; Wei, W.; Li, X.; Shen, P.; Ju, D.; Wang, Z.; Zhang, R.; Yang, F.; Chen, C.; Cao, K.; et al. BMI1 and MEL18 Promote Colitis-Associated Cancer in Mice via REG3B and STAT3. Gastroenterology 2017, 153, 1607–1620. [Google Scholar] [CrossRef]
  70. Ye, Y.; Xiao, L.; Wang, S.-J.; Yue, W.; Yin, Q.-S.; Sun, M.-Y.; Xia, W.; Shao, Z.-Y.; Zhang, H. Up-Regulation of REG3A in Colorectal Cancer Cells Confers Proliferation and Correlates with Colorectal Cancer Risk. Oncotarget 2016, 7, 3921–3933. [Google Scholar] [CrossRef]
  71. Qiu, Y.-S.; Liao, G.-J.; Jiang, N.-N. REG3A Overexpression Suppresses Gastric Cancer Cell Invasion, Proliferation and Promotes Apoptosis through PI3K/Akt Signaling Pathway. Int. J. Mol. Med. 2018, 41, 3167–3174. [Google Scholar] [CrossRef]
  72. Chen, Z.-F.; Huang, Z.-M.; Xue, H.-B.; Lin, X.-Q.; Chen, R.-P.; Chen, M.-J.; Jin, R.-F. REG3A Promotes the Proliferation, Migration, and Invasion of Gastric Cancer Cells. Onco. Targets Ther. 2017, 10, 2017–2023. [Google Scholar] [CrossRef]
  73. Yuan, R.-H.; Jeng, Y.-M.; Chen, H.-L.; Hsieh, F.-J.; Yang, C.-Y.; Lee, P.-H.; Hsu, H.-C. Opposite Roles of Human Pancreatitis-Associated Protein and REG1A Expression in Hepatocellular Carcinoma: Association of Pancreatitis-Associated Protein Expression with Low-Stage Hepatocellular Carcinoma, Beta-Catenin Mutation, and Favorable Prognosis. Clin. Cancer Res. 2005, 11, 2568–2575. [Google Scholar] [CrossRef]
  74. Cho, Y.; Park, M.J.; Kim, K.; Park, J.-Y.; Kim, J.; Kim, W.; Yoon, J.-H. Tumor-Stroma Crosstalk Enhances REG3A Expressions That Drive the Progression of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2020, 21, 472. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, X.; Wang, J.; Wang, H.; Yin, G.; Liu, Y.; Lei, X.; Xiang, M. REG3A Accelerates Pancreatic Cancer Cell Growth under IL-6-Associated Inflammatory Condition: Involvement of a REG3A-JAK2/STAT3 Positive Feedback Loop. Cancer Lett. 2015, 362, 45–60. [Google Scholar] [CrossRef] [PubMed]
  76. Loncle, C.; Bonjoch, L.; Folch-Puy, E.; Lopez-Millan, M.B.; Lac, S.; Molejon, M.I.; Chuluyan, E.; Cordelier, P.; Dubus, P.; Lomberk, G.; et al. IL17 Functions through the Novel REG3β-JAK2-STAT3 Inflammatory Pathway to Promote the Transition from Chronic Pancreatitis to Pancreatic Cancer. Cancer Res. 2015, 75, 4852–4862. [Google Scholar] [CrossRef] [PubMed]
  77. Ren, X.; Teng, Y.; Xie, K.; He, X.; Chen, G.; Zhang, K.; Liao, Q.; Zhang, J.; Zhou, X.; Zhu, Y.; et al. REG3A Secreted by Peritumoral Acinar Cells Enhances Pancreatic Ductal Adenocarcinoma Progression via Activation of EGFR Signaling. Cell Commun. Signal. 2025, 23, 96. [Google Scholar] [CrossRef]
  78. Liu, X.; Zhou, Z.; Cheng, Q.; Wang, H.; Cao, H.; Xu, Q.; Tuo, Y.; Jiang, L.; Zou, Y.; Ren, H.; et al. Acceleration of Pancreatic Tumorigenesis under Immunosuppressive Microenvironment Induced by Reg3g Overexpression. Cell Death Dis. 2017, 8, e3033. [Google Scholar] [CrossRef]
  79. Zhang, H.; Corredor, A.L.G.; Messina-Pacheco, J.; Li, Q.; Zogopoulos, G.; Kaddour, N.; Wang, Y.; Shi, B.-Y.; Gregorieff, A.; Liu, J.-L.; et al. REG3A/REG3B Promotes Acinar to Ductal Metaplasia through Binding to EXTL3 and Activating the RAS-RAF-MEK-ERK Signaling Pathway. Commun. Biol. 2021, 4, 688. [Google Scholar] [CrossRef]
  80. Yin, G.; Du, J.; Cao, H.; Liu, X.; Xu, Q.; Xiang, M. Reg3g Promotes Pancreatic Carcinogenesis in a Murine Model of Chronic Pancreatitis. Dig. Dis. Sci. 2015, 60, 3656–3668. [Google Scholar] [CrossRef]
  81. Mukherjee, S.; Zheng, H.; Derebe, M.G.; Callenberg, K.M.; Partch, C.L.; Rollins, D.; Propheter, D.C.; Rizo, J.; Grabe, M.; Jiang, Q.-X.; et al. Antibacterial Membrane Attack by a Pore-Forming Intestinal C-Type Lectin. Nature 2014, 505, 103–107. [Google Scholar] [CrossRef]
  82. Mukherjee, S.; Vaishnava, S.; Hooper, L.V. Multi-Layered Regulation of Intestinal Antimicrobial Defense. Cell. Mol. Life Sci. 2008, 65, 3019–3027. [Google Scholar] [CrossRef]
  83. Stelter, C.; Käppeli, R.; König, C.; Krah, A.; Hardt, W.-D.; Stecher, B.; Bumann, D. Salmonella-Induced Mucosal Lectin RegIIIβ Kills Competing Gut Microbiota. PLoS ONE 2011, 6, e20749. [Google Scholar] [CrossRef]
  84. Miki, T.; Holst, O.; Hardt, W.-D. The Bactericidal Activity of the C-Type Lectin RegIIIβ against Gram-Negative Bacteria Involves Binding to Lipid A. J. Biol. Chem. 2012, 287, 34844–34855. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, L.; Fouts, D.E.; Stärkel, P.; Hartmann, P.; Chen, P.; Llorente, C.; DePew, J.; Moncera, K.; Ho, S.B.; Brenner, D.A.; et al. Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation. Cell Host Microbe 2016, 19, 227–239. [Google Scholar] [CrossRef] [PubMed]
  86. Darnaud, M.; Dos Santos, A.; Gonzalez, P.; Augui, S.; Lacoste, C.; Desterke, C.; De Hertogh, G.; Valentino, E.; Braun, E.; Zheng, J.; et al. Enteric Delivery of Regenerating Family Member 3 Alpha Alters the Intestinal Microbiota and Controls Inflammation in Mice with Colitis. Gastroenterology 2018, 154, 1009–1023.e14. [Google Scholar] [CrossRef] [PubMed]
  87. Frazier, K.; Kambal, A.; Zale, E.A.; Pierre, J.F.; Hubert, N.; Miyoshi, S.; Miyoshi, J.; Ringus, D.L.; Harris, D.; Yang, K.; et al. High-Fat Diet Disrupts REG3γ and Gut Microbial Rhythms Promoting Metabolic Dysfunction. Cell Host Microbe 2022, 30, 809–823.e6. [Google Scholar] [CrossRef]
  88. Nalpas, B.; Ichaï, P.; Jamot, L.; Carbonell, N.; Rudler, M.; Mathurin, P.; Durand, F.; Gerken, G.; Manns, M.; Trautwein, C.; et al. A Proof of Concept, Phase II Randomized European Trial, on the Efficacy of ALF-5755, a Novel Extracellular Matrix-Targeted Antioxidant in Patients with Acute Liver Diseases. PLoS ONE 2016, 11, e0150733. [Google Scholar] [CrossRef]
  89. Simon, M.-T.; Pauloin, A.; Normand, G.; Lieu, H.-T.; Mouly, H.; Pivert, G.; Carnot, F.; Tralhao, J.G.; Brechot, C.; Christa, L. HIP/PAP Stimulates Liver Regeneration after Partial Hepatectomy and Combines Mitogenic and Anti-Apoptotic Functions through the PKA Signaling Pathway. FASEB J. 2003, 17, 1441–1450. [Google Scholar] [CrossRef]
  90. Lieu, H.-T.; Batteux, F.; Simon, M.-T.; Cortes, A.; Nicco, C.; Zavala, F.; Pauloin, A.; Tralhao, J.G.; Soubrane, O.; Weill, B.; et al. HIP/PAP Accelerates Liver Regeneration and Protects against Acetaminophen Injury in Mice. Hepatology 2005, 42, 618–626. [Google Scholar] [CrossRef]
  91. Lieu, H.-T.; Simon, M.-T.; Nguyen-Khoa, T.; Kebede, M.; Cortes, A.; Tebar, L.; Smith, A.J.H.; Bayne, R.; Hunt, S.P.; Bréchot, C.; et al. Reg2 Inactivation Increases Sensitivity to Fas Hepatotoxicity and Delays Liver Regeneration Post-Hepatectomy in Mice. Hepatology 2006, 44, 1452–1464. [Google Scholar] [CrossRef] [PubMed]
  92. Haldipur, P.; Dupuis, N.; Degos, V.; Moniaux, N.; Chhor, V.; Rasika, S.; Schwendimann, L.; le Charpentier, T.; Rougier, E.; Amouyal, P.; et al. HIP/PAP Prevents Excitotoxic Neuronal Death and Promotes Plasticity. Ann. Clin. Transl. Neurol. 2014, 1, 739–754. [Google Scholar] [CrossRef] [PubMed]
  93. Nishimune, H.; Vasseur, S.; Wiese, S.; Birling, M.C.; Holtmann, B.; Sendtner, M.; Iovanna, J.L.; Henderson, C.E. Reg-2 Is a Motoneuron Neurotrophic Factor and a Signalling Intermediate in the CNTF Survival Pathway. Nat. Cell Biol. 2000, 2, 906–914. [Google Scholar] [CrossRef] [PubMed]
  94. Namikawa, K.; Fukushima, M.; Murakami, K.; Suzuki, A.; Takasawa, S.; Okamoto, H.; Kiyama, H. Expression of Reg/PAP Family Members during Motor Nerve Regeneration in Rat. Biochem. Biophys. Res. Commun. 2005, 332, 126–134. [Google Scholar] [CrossRef]
  95. Lörchner, H.; Pöling, J.; Gajawada, P.; Hou, Y.; Polyakova, V.; Kostin, S.; Adrian-Segarra, J.M.; Boettger, T.; Wietelmann, A.; Warnecke, H.; et al. Myocardial Healing Requires Reg3β-Dependent Accumulation of Macrophages in the Ischemic Heart. Nat. Med. 2015, 21, 353–362. [Google Scholar] [CrossRef]
  96. Lörchner, H.; Hou, Y.; Adrian-Segarra, J.M.; Kulhei, J.; Detzer, J.; Günther, S.; Gajawada, P.; Warnecke, H.; Niessen, H.W.; Pöling, J.; et al. Reg Proteins Direct Accumulation of Functionally Distinct Macrophage Subsets after Myocardial Infarction. Cardiovasc. Res. 2018, 114, 1667–1679. [Google Scholar] [CrossRef]
  97. Kim, B.T.; Kitagawa, H.; Tamura, J.; Saito, T.; Kusche-Gullberg, M.; Lindahl, U.; Sugahara, K. Human Tumor Suppressor EXT Gene Family Members EXTL1 and EXTL3 Encode Alpha 1,4- N-Acetylglucosaminyltransferases That Likely Are Involved in Heparan Sulfate/ Heparin Biosynthesis. Proc. Natl. Acad. Sci. USA 2001, 98, 7176–7181. [Google Scholar] [CrossRef]
  98. Kobayashi, S.; Akiyama, T.; Nata, K.; Abe, M.; Tajima, M.; Shervani, N.J.; Unno, M.; Matsuno, S.; Sasaki, H.; Takasawa, S.; et al. Identification of a Receptor for Reg (Regenerating Gene) Protein, a Pancreatic Beta-Cell Regeneration Factor. J. Biol. Chem. 2000, 275, 10723–10726. [Google Scholar] [CrossRef]
  99. Levetan, C.S.; Upham, L.V.; Deng, S.; Laury-Kleintop, L.; Kery, V.; Nolan, R.; Quinlan, J.; Torres, C.; El-Hajj, R.J. Discovery of a Human Peptide Sequence Signaling Islet Neogenesis. Endocr. Pract. 2008, 14, 1075–1083. [Google Scholar] [CrossRef]
  100. Takasawa, S.; Ikeda, T.; Akiyama, T.; Nata, K.; Nakagawa, K.; Shervani, N.J.; Noguchi, N.; Murakami-Kawaguchi, S.; Yamauchi, A.; Takahashi, I.; et al. Cyclin D1 Activation through ATF-2 in Reg-Induced Pancreatic Beta-Cell Regeneration. FEBS Lett. 2006, 580, 585–591. [Google Scholar] [CrossRef]
  101. Watanabe, T.; Yonemura, Y.; Yonekura, H.; Suzuki, Y.; Miyashita, H.; Sugiyama, K.; Moriizumi, S.; Unno, M.; Tanaka, O.; Kondo, H. Pancreatic Beta-Cell Replication and Amelioration of Surgical Diabetes by Reg Protein. Proc. Natl. Acad. Sci. USA 1994, 91, 3589–3592. [Google Scholar] [CrossRef]
  102. Petropavlovskaia, M.; Daoud, J.; Zhu, J.; Moosavi, M.; Ding, J.; Makhlin, J.; Assouline-Thomas, B.; Rosenberg, L. Mechanisms of Action of Islet Neogenesis-Associated Protein: Comparison of the Full-Length Recombinant Protein and a Bioactive Peptide. Am. J. Physiol. Endocrinol. Metab. 2012, 303, E917–E927. [Google Scholar] [CrossRef]
  103. Jamal, A.-M.; Lipsett, M.; Sladek, R.; Laganière, S.; Hanley, S.; Rosenberg, L. Morphogenetic Plasticity of Adult Human Pancreatic Islets of Langerhans. Cell Death Differ. 2005, 12, 702–712. [Google Scholar] [CrossRef]
  104. Lipsett, M.A.; Austin, E.B.; Castellarin, M.L.; Lemay, J.; Rosenberg, L. Evidence for the Homeostatic Regulation of Induced Beta Cell Mass Expansion. Diabetologia 2006, 49, 2910–2919. [Google Scholar] [CrossRef] [PubMed]
  105. Kapur, R.; Højfeldt, T.W.; Højfeldt, T.W.; Rønn, S.G.; Karlsen, A.E.; Heller, R.S. Short-Term Effects of INGAP and Reg Family Peptides on the Appearance of Small β-Cells Clusters in Non-Diabetic Mice. Islets 2012, 4, 40–48. [Google Scholar] [CrossRef] [PubMed]
  106. Dungan, K.M.; Buse, J.B.; Ratner, R.E. Effects of Therapy in Type 1 and Type 2 Diabetes Mellitus with a Peptide Derived from Islet Neogenesis Associated Protein (INGAP). Diabetes Metab. Res. Rev. 2009, 25, 558–565. [Google Scholar] [CrossRef] [PubMed]
  107. Xiong, X.; Wang, X.; Li, B.; Chowdhury, S.; Lu, Y.; Srikant, C.B.; Ning, G.; Liu, J.-L. Pancreatic Islet-Specific Overexpression of Reg3β Protein Induced the Expression of pro-Islet Genes and Protected the Mice against Streptozotocin-Induced Diabetes Mellitus. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E669–E680. [Google Scholar] [CrossRef]
  108. Huang, Y.; Qi, H.; Zhang, Z.; Wang, E.; Yun, H.; Yan, H.; Su, X.; Liu, Y.; Tang, Z.; Gao, Y.; et al. Gut REG3γ-Associated Lactobacillus Induces Anti-Inflammatory Macrophages to Maintain Adipose Tissue Homeostasis. Front. Immunol. 2017, 8, 1063. [Google Scholar] [CrossRef] [PubMed]
  109. Hartmann, P.; Seebauer, C.T.; Mazagova, M.; Horvath, A.; Wang, L.; Llorente, C.; Varki, N.M.; Brandl, K.; Ho, S.B.; Schnabl, B. Deficiency of Intestinal Mucin-2 Protects Mice from Diet-Induced Fatty Liver Disease and Obesity. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G310–G322. [Google Scholar] [CrossRef]
  110. Bluemel, S.; Wang, L.; Martino, C.; Lee, S.; Wang, Y.; Williams, B.; Horvath, A.; Stadlbauer, V.; Zengler, K.; Schnabl, B. The Role of Intestinal C-Type Regenerating Islet Derived-3 Lectins for Nonalcoholic Steatohepatitis. Hepatol. Commun. 2018, 2, 393–406. [Google Scholar] [CrossRef]
  111. Montalto, G.; Iovanna, J.L.; Soresi, M.; Dusetti, N.; Carroccio, A.; Barthelemy-Bialas, S.; Cartabellotta, A.; Dagorn, J.C. Clinical Evaluation of Pancreatitis-Associated Protein as a Serum Marker of Hepatocellular Carcinoma: Comparison with Alpha-Fetoprotein. Oncology 1998, 55, 421–425. [Google Scholar] [CrossRef]
  112. Wang, J.; Zhou, H.; Han, Y.; Liu, X.; Wang, M.; Wang, X.; Yin, G.; Li, X.; Xiang, M. SOCS3 Methylation in Synergy with Reg3A Overexpression Promotes Cell Growth in Pancreatic Cancer. J. Mol. Med. 2014, 92, 1257–1269. [Google Scholar] [CrossRef]
  113. Zhang, P.; Zhang, J.; Yu, M.; Zhang, X.; Li, H. Over-Expression of REG 3 A Gene Can Inhibit the Proliferation, Invasion and Migration of Human Breast Cancer. Int. J. Clin. Exp. Pathol. 2016, 9, 12104–12113. [Google Scholar]
  114. Rosty, C.; Christa, L.; Kuzdzal, S.; Baldwin, W.M.; Zahurak, M.L.; Carnot, F.; Chan, D.W.; Canto, M.; Lillemoe, K.D.; Cameron, J.L.; et al. Identification of Hepatocarcinoma-Intestine-Pancreas/Pancreatitis-Associated Protein I as a Biomarker for Pancreatic Ductal Adenocarcinoma by Protein Biochip Technology. Cancer Res. 2002, 62, 1868–1875. [Google Scholar] [PubMed]
  115. Cerwenka, H.; Aigner, R.; Bacher, H.; Werkgartner, G.; el-Shabrawi, A.; Quehenberger, F.; Mischinger, H.J. Pancreatitis-Associated Protein (PAP) in Patients with Pancreatic Cancer. Anticancer Res. 2001, 21, 1471–1474. [Google Scholar] [PubMed]
  116. Xu, Q.; Fu, R.; Yin, G.; Liu, X.; Liu, Y.; Xiang, M. Microarray-Based Gene Expression Profiling Reveals Genes and Pathways Involved in the Oncogenic Function of REG3A on Pancreatic Cancer Cells. Gene 2016, 578, 263–273. [Google Scholar] [CrossRef]
  117. Fukushima, N.; Koopmann, J.; Sato, N.; Prasad, N.; Carvalho, R.; Leach, S.D.; Hruban, R.H.; Goggins, M. Gene Expression Alterations in the Non-Neoplastic Parenchyma Adjacent to Infiltrating Pancreatic Ductal Adenocarcinoma. Mod. Pathol. 2005, 18, 779–787. [Google Scholar] [CrossRef]
  118. Wang, G.; Zhao, X.; Li, D.-C. Expression of HIP/PAP in Hepatocellular Carcinoma and Effect of siRNA on Migration and Invasion in HCC Cells. Asian Pac. J. Trop. Med. 2015, 8, 848–854. [Google Scholar] [CrossRef]
  119. Kimura, M.; Naito, H.; Tojo, T.; Itaya-Hironaka, A.; Dohi, Y.; Yoshimura, M.; Nakagawara, K.-I.; Takasawa, S.; Taniguchi, S. REG Iα Gene Expression Is Linked with the Poor Prognosis of Lung Adenocarcinoma and Squamous Cell Carcinoma Patients via Discrete Mechanisms. Oncol. Rep. 2013, 30, 2625–2631. [Google Scholar] [CrossRef]
  120. Mikami, S.; Ota, I.; Masui, T.; Uchiyama, T.; Okamoto, H.; Kimura, T.; Takasawa, S.; Kitahara, T. Resveratrol-induced REG III Expression Enhances Chemo- and Radiosensitivity in Head and Neck Cancer in Xenograft Mice. Oncol. Rep. 2019, 42, 436–442. [Google Scholar] [CrossRef]
  121. Wang, L.; Tuo, H.; Song, Z.; Li, W.; Peng, Y. Reg3A (Regenerating Family Member 3 Alpha) Acts as a Tumor Suppressor by Targeting DMBT1 (Deleted in Malignant Brain Tumors 1) in Gastric Cancer. Bioengineered 2021, 12, 7644–7655. [Google Scholar] [CrossRef]
  122. Yari, H.; Jin, L.; Teng, L.; Wang, Y.; Wu, Y.; Liu, G.Z.; Gao, W.; Liang, J.; Xi, Y.; Feng, Y.C.; et al. LncRNA REG1CP Promotes Tumorigenesis through an Enhancer Complex to Recruit FANCJ Helicase for REG3A Transcription. Nat. Commun. 2019, 10, 5334. [Google Scholar] [CrossRef]
  123. Fernandez, E.; Wargo, J.A.; Helmink, B.A. The Microbiome and Cancer: A Translational Science Review. JAMA 2025, 333, 2188–2196. [Google Scholar] [CrossRef]
  124. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium Nucleatum Potentiates Intestinal Tumorigenesis and Modulates the Tumor-Immune Microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [PubMed]
  125. Permain, J.; Hock, B.; Eglinton, T.; Purcell, R. Functional Links between the Microbiome and the Molecular Pathways of Colorectal Carcinogenesis. Cancer Metastasis Rev. 2024, 43, 1463–1474. [Google Scholar] [CrossRef] [PubMed]
  126. Luo, M.; Li, Q.; Gu, Q.; Zhang, C. Fusobacterium Nucleatum: A Novel Regulator of Antitumor Immune Checkpoint Blockade Therapy in Colorectal Cancer. Am. J. Cancer Res. 2024, 14, 3962–3975. [Google Scholar] [CrossRef]
  127. Li, S.; Duan, Y.; Luo, S.; Zhou, F.; Wu, Q.; Lu, Z. Short-Chain Fatty Acids and Cancer. Trends Cancer 2025, 11, 154–168. [Google Scholar] [CrossRef]
  128. Zhou, X.; Wang, L.-Q.; Song, S.; Xu, M.; Li, C.-P. Helicobacter Pylori Infection Promotes the Progression of Gastric Cancer by Regulating the Expression of DMBT1. World J. Clin. Oncol. 2025, 16, 105322. [Google Scholar] [CrossRef]
  129. Lee, K.S.; Kalantzis, A.; Jackson, C.B.; O’Connor, L.; Murata-Kamiya, N.; Hatakeyama, M.; Judd, L.M.; Giraud, A.S.; Menheniott, T.R. Helicobacter Pylori CagA Triggers Expression of the Bactericidal Lectin REG3γ via Gastric STAT3 Activation. PLoS ONE 2012, 7, e30786. [Google Scholar] [CrossRef]
Figure 1. Domain structure of human REG3A. Schematic representation of the secondary structure of human REG3A (amino acids 1–175), highlighting the cleavage sites for trypsin and elastase, loops 1 and 2, and the three disulfide bridges. The green box denotes the mature REG3A lectin domain, which contains a single carbohydrate recognition domain (CRD). Cysteine residues involved in disulfide bonding are indicated by the letter C within circles. (Created in BioRender. https://BioRender.com/7r8q7cv (accessed on 12 June 2025).
Figure 1. Domain structure of human REG3A. Schematic representation of the secondary structure of human REG3A (amino acids 1–175), highlighting the cleavage sites for trypsin and elastase, loops 1 and 2, and the three disulfide bridges. The green box denotes the mature REG3A lectin domain, which contains a single carbohydrate recognition domain (CRD). Cysteine residues involved in disulfide bonding are indicated by the letter C within circles. (Created in BioRender. https://BioRender.com/7r8q7cv (accessed on 12 June 2025).
Cancers 17 02395 g001
Figure 2. Dual antimicrobial and antioxidant functions of REG3A in the gut. Intestinal bacteria are shown in pink (bacilli) and brown (cocci). ROS: reactive oxygen species. (Created in BioRender. https://BioRender.com/nrtonjp (accessed on 16 June 2025).
Figure 2. Dual antimicrobial and antioxidant functions of REG3A in the gut. Intestinal bacteria are shown in pink (bacilli) and brown (cocci). ROS: reactive oxygen species. (Created in BioRender. https://BioRender.com/nrtonjp (accessed on 16 June 2025).
Cancers 17 02395 g002
Figure 3. Context-dependent functions of REG3A in cancer. This schematic summarizes the dual roles of REG3A in cancer as an oncogene in pancreatic ductal adenocarcinoma (PDAC) and as a tumor suppressor in hepatocellular carcinoma (HCC). In PDAC, REG3A is induced by IL-6/JAK/STAT signaling in inflamed acinar cells (pink) adjacent to tumor lesions (green), promoting tumor progression via EGFR (and possibly EXTL3) activation and downstream pathways. In HCC, REG3A is expressed in early-stage hepatocytes, where it interacts with hexosamine biosynthesis pathway intermediates, lowering UDP-GlcNAc levels and altering metabolism to delay tumor development. (Created in BioRender. https://BioRender.com/lf76afg (accessed on 12 June 2025).
Figure 3. Context-dependent functions of REG3A in cancer. This schematic summarizes the dual roles of REG3A in cancer as an oncogene in pancreatic ductal adenocarcinoma (PDAC) and as a tumor suppressor in hepatocellular carcinoma (HCC). In PDAC, REG3A is induced by IL-6/JAK/STAT signaling in inflamed acinar cells (pink) adjacent to tumor lesions (green), promoting tumor progression via EGFR (and possibly EXTL3) activation and downstream pathways. In HCC, REG3A is expressed in early-stage hepatocytes, where it interacts with hexosamine biosynthesis pathway intermediates, lowering UDP-GlcNAc levels and altering metabolism to delay tumor development. (Created in BioRender. https://BioRender.com/lf76afg (accessed on 12 June 2025).
Cancers 17 02395 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Faivre, J.; Shalhoub, H.; Nguyen, T.S.; Xie, H.; Moniaux, N. REG3A: A Multifunctional Antioxidant Lectin at the Crossroads of Microbiota Regulation, Inflammation, and Cancer. Cancers 2025, 17, 2395. https://doi.org/10.3390/cancers17142395

AMA Style

Faivre J, Shalhoub H, Nguyen TS, Xie H, Moniaux N. REG3A: A Multifunctional Antioxidant Lectin at the Crossroads of Microbiota Regulation, Inflammation, and Cancer. Cancers. 2025; 17(14):2395. https://doi.org/10.3390/cancers17142395

Chicago/Turabian Style

Faivre, Jamila, Hala Shalhoub, Tung Son Nguyen, Haishen Xie, and Nicolas Moniaux. 2025. "REG3A: A Multifunctional Antioxidant Lectin at the Crossroads of Microbiota Regulation, Inflammation, and Cancer" Cancers 17, no. 14: 2395. https://doi.org/10.3390/cancers17142395

APA Style

Faivre, J., Shalhoub, H., Nguyen, T. S., Xie, H., & Moniaux, N. (2025). REG3A: A Multifunctional Antioxidant Lectin at the Crossroads of Microbiota Regulation, Inflammation, and Cancer. Cancers, 17(14), 2395. https://doi.org/10.3390/cancers17142395

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop