The intestinal mucosal barrier is a critical component of the gastrointestinal tract, serving as the front-line defense against pathogens, toxins, and dietary antigens. Its integrity is paramount to health [1]. Inversely, dysfunction of barrier integrity contributes to inflammatory bowel disease (IBD), neurodegenerative diseases, and systemic metabolic dysfunction [2,3]. In recent years, a brush-border metalloenzyme, intestinal alkaline phosphatase (ALPi), has emerged as a pivotal regulator of gut homeostasis and a promising therapeutic target.
This article summarizes the biological functions of ALPi, its mechanism of action in protecting the gut mucosal barrier, its implications in health and disease, and its potential as a biomarker or therapeutic tool.
Table of Contents
2. Mechanisms of ALPi in Protecting the Intestinal Mucosal Barrier
3. ALPi and Disease Associations
ALPi, also known as IAP, is a brush border enzyme primarily produced by enterocytes (that line the small intestine) and then secreted into the lumen, blood, and stool.
ALPi hydrolyzes phosphate from the lipid A moiety of lipopolysaccharides (LPS) and thus significantly reduces LPS pro-inflammatory activity [4]. In addition to detoxifying LPS, ALPi also protects gut barrier integrity, regulates gut microbiota, dephosphorylates pro-inflammatory nucleotides, and modulates bicarbonate secretion and duodenal surface pH [4-6]. ALPi is essential for intestinal homeostasis and microbial balance.
ALPi plays a critical role in maintaining the integrity and functionality of the intestinal mucosal barrier. Its mechanisms of action are multifaceted, mainly involving regulation of tight junction proteins, LPS detoxification, attenuation of inflammation, and maintenance of gut microbial homeostasis.
The intestinal mucosal barrier relies on tight junction proteins (TJPs) such as ZO-1, ZO-2, Occludin, and Claudins to maintain epithelial integrity. These TJPs regulate paracellular permeability and prevent the translocation of harmful substances. ALPi upregulates the expression of these tight junction proteins (TJPs), thus maintaining tight junction integrity and reducing gut permeability (leaky gut) [8]. Additionally, IAP preserves the localization of these proteins during inflammation, further strengthening barrier function [5].
Animal studies demonstrated that ALPi deficiency impairs the barrier. In necrotizing enterocolitis (NEC) rat models, exogenous bovine ALPi reduced permeability by up to 15-fold [7]. Knockout mice lacking ALPi had reduced expression of TJPs, including ZO-1, ZO-2, and occludin, and claudin 1 when compared to wild-type mice [9]. The administration of exogenous ALPi supplementation was demonstrated to prevent alcohol-induced intestinal barrier dysfunction and TJP loss [10].
ALPi also alleviates inflammation-caused damage to TJPs by detoxifying lipopolysaccharides (LPS) and reducing pro-inflammatory cytokine levels. This protective effect helps preserve tight junction structure and function [11,12].
ALPi is a key enzyme that detoxifies LPS, a pro-inflammatory component of Gram-negative bacteria. ALPi detoxifies LPS, thereby inhibiting downstream activation of immunocytes (TNF-α, IL-1β, IL-6, IL-12) and the subsequent inflammatory responses through TLR4 signaling [13]. Hwang et al. also indicated that ALPi maintains the normal function of the intestinal mucosal barrier through the TLR4/NF-κB-mediated pathway [14].
Figure 1. Detoxification of LPS by ALPi
The picture is cited from:https://pmc.ncbi.nlm.nih.gov/articles/PMC9026380/
LPS detoxification is critical to prevent endotoxemia, a condition where LPS translocates from the gut into the bloodstream, causing systemic inflammation and organ dysfunction [15].
During inflammation, extracellular purines (e.g., adenosine, ADP, ATP) and pyrimidines (e.g., UDP, UTP) released from host cells and gut microbiota activate purinergic receptors autocrinally/paracrinally [16]. Extracellular ATP is hydrolyzed by ALPi to ADP, AMP, and adenosine [17], which then binds adenosine receptors (A1, A2A, A2B, A3) to provide intestinal protection and anti-inflammatory effects [18]. M. Estaki et al. showed that oral ALPi alleviates metabolic syndrome in ALPi-knockout mice fed a high-fat diet [19].
ALPi exerts anti-inflammatory effects by dephosphorylating pro-inflammatory molecules like ATP and flagellin [17], thereby reducing activation of inflammatory pathways such as the NF-κB signaling pathway, which are involved in gut barrier dysfunction [6]. Orally administered ALPi has been shown to decrease levels of pro-inflammatory cytokines, including TNF-α, IL-1β, and IL-8, thus alleviating intestinal inflammation [12][17].
ALPi activity is closely tied to microbiota composition. In murine models, ALPi deficiency leads to dysbiosis characterized by an overgrowth of harmful bacteria [21]. Conversely, oral ALPi supplementation restores microbial balance and ameliorates colitis symptoms [8]. By shaping microbial ecology, ALPi helps maintain symbiosis and prevents the cascade of chronic intestinal inflammation.
ALPi helps maintain gut microbial homeostasis by promoting mucosal tolerance to gut bacteria through LPS detoxification [15]. ALPi-knockout mice show increased bacterial translocation after intestinal injury [20] and reduced bacterial diversity, reversible with ALPi supplementation [21]. ALPi supports bacterial growth by dephosphorylating luminal ATP (which inhibits Gram-positive bacteria) [22,23]. Exogenous ALPi administration alleviates Salmonella and C. difficile infections by restoring normal gut microbiota [24].
Additionally, ALPi functions as an alkaline sensor in the duodenum, regulating bicarbonate secretion and mucosal pH. Optimal ALPi activity occurs at mucosal pH >8. Acidic environments reduce its enzymatic function, thereby impairing the intestinal mucosal barrier [25]. Thus, ALPi contributes to maintaining a balanced pH, supporting microbial homeostasis and mucosal protection.
ALPi is a critical enzyme that plays a significant role in maintaining intestinal homeostasis and barrier function. Its deficiency or altered activity has been implicated in various intestinal diseases, ranging from inflammatory conditions to dysbiosis and increased permeability.
IBD pathogenesis involves aberrant immune responses to gut bacteria. Studies have shown that ALPi deficiency or reduced activity is linked to increased intestinal inflammation, dysbiosis, and systemic inflammation, all of which are hallmarks of IBD [26]. ALPi detoxifies LPS, enhances autophagy in epithelial cells and macrophages, and reduces permeability. Oral administration of ALPi significantly alleviated colitis severity in mouse IBD models [27].
NEC is a severe gastrointestinal condition primarily affecting preterm infants, often leading to high morbidity and mortality rates. LPS is a key mediator of inflammation in NEC. Studies have shown that preterm infants with NEC exhibit significantly lower ALPi levels compared to healthy controls, suggesting that ALPi deficiency may contribute to the development of NEC [28].
Additionally, ALPi has been found to promote autophagy, improve intestinal perfusion, and prevent dysbiosis, further protecting against NEC-related intestinal injury. These findings underscore the enzyme's potential as a preventive and therapeutic agent in NEC management [29,30].
Sepsis-induced AKI involves systemic inflammation and LPS toxicity. Clinical trials demonstrated that intravenous recombinant ALPi improved renal function, reduced plasma creatinine, and shortened dialysis duration in AKI patients [31].
ALPi-deficient mice exhibit dysbiosis, type 2 diabetes, and hyperlipidemia. Oral ALPi restores microbial balance and protects against metabolic syndrome [32]. Overexpression of ALPi in animal models preserved the mucin layer, reduced LPS absorption, and decreased atherosclerosis progression [33]. These findings suggest ALPi plays a crucial role in metabolic health.
Recent Gut and JCI publications highlight recombinant ALPi as a therapeutic candidate for multiple diseases, including IBD, AKI, and metabolic syndrome. AM-Pharma has advanced recombinant human ALPi (recAP) into clinical trials, showing safety and efficacy in sepsis-related AKI [31]. Furthermore, the role of ALPi in microbiome regulation is an emerging area, connecting gut health to systemic diseases such as neuroinflammation and cardiovascular disorders.
Specific antibodies, recombinant ALPi proteins, and ELISA kits are essential to support ongoing research. These reagents enable:
CUSABIO has developed a comprehensive portfolio of ALPi-related products, including recombinant ALPi proteins, ALPi polyclonal antibodies, and sensitive ALPi ELISA kits. These reagents provide reliable tools for exploring ALPi function, signaling pathways, and therapeutic potential.
ALPi stands as a critical enzymatic defender of the gut mucosal barrier, safeguarding intestinal integrity by detoxifying endotoxins, regulating immune responses, and preserving tight junction function. Its relevance in diverse inflammatory and metabolic pathologies underscores its promising potential as a biomarker and therapeutic agent. Cutting-edge ALPi antibodies, recombinant proteins, and ELISA kits now empower researchers to dissect its role with precision and accelerate discoveries toward novel gut-targeted interventions.
References
[1] Vancamelbeke, M.; Vermeire, S. The intestinal barrier: A fundamental role in health and disease [J]. Expert. Rev. Gastroenterol. Hepatol. 2017, 11, 821–834.
[2] Metta, V.; Leta, V.; et al. Gastrointestinal dysfunction in Parkinson’s disease: Molecular pathology and implications of gut microbiome, probiotics, and fecal microbiota transplantation [J]. J. Neurol. 2021. [Google Scholar] [CrossRef]
[3] DeGruttola, A.K.; Low, D.; et al. Current Understanding of Dysbiosis in Disease in Human and Animal Models [J]. Inflamm. Bowel. Dis. 2016, 22, 1137–1150.
[4] Beumer, C.; Wulferink, M.; et al. Calf intestinal alkaline phosphatase, a novel therapeutic drug for lipopolysaccharide (LPS)-mediated diseases, attenuates LPS toxicity in mice and piglets [J]. J. Pharmacol. Exp. Ther. 2003, 307, 737–744.
[5] Liu, W.; Hu, D.; et al. Intestinal Alkaline Phosphatase Regulates Tight Junction Protein Levels [J]. J. Am. Coll. Surg. 2016, 222, 1009–1017.
[6] Lalles, J.P. Intestinal alkaline phosphatase: Novel functions and protective effects [J]. Nutr. Rev. 2014, 72, 82–94.
[7] Rentea RM, Liedel JL, et al. Intestinal alkaline phosphatase administration in newborns is protective of gut barrier function in necrotizing enterocolitis [J]. J Pediatr Surg. 2012;47(6):1135–1142.
[8] Chen KT, Malo MS, et al. A role for intestinal alkaline phosphatase in the maintenance of local gut immunity [J].Digestive Diseases and Sciences, 56(4), 1020.
[9] Hamarneh SR, Mohamed MM, et al. A novel approach to maintain gut mucosal integrity using an oral enzyme supplement [J]. Ann Surg 2014; 260: 706-14.
[10] Hamarneh, S.R.; Kim, B.M.; et al. Intestinal Alkaline Phosphatase Attenuates Alcohol-Induced Hepatosteatosis in Mice [J]. Dig. Dis. Sci. 2017, 62, 2021–2034.
[11] Dissanayake WMN, Chandanee MR, et al. Change in intestinal alkaline phosphatase activity is a hallmark of antibiotic-induced intestinal dysbiosis [J]. Anim Biosci. 2023 Sep;36(9):1403-1413.
[12] Luo Z, Wang Z, et al. Intestinal alkaline phosphatase improves intestinal permeability and alleviates multiple organ dysfunction caused by heatstroke [J]. Heliyon. 2023 Nov 4;9(11):e21838.
[13] Bentala H, Verweij WR, et al. Removal of phosphate from lipid A as a strategy to detoxify lipopolysaccharide [J]. Shock. 2002;18(6):561–566.
[14] Cario E. Bacterial interactions with cells of the intestinal mucosa [J]. toll-like receptors and NOD2. Gut, 2005, 54( 8): 1182-1193.
[15] Bates JM, Akerlund J, Mittge E, Guillemin K. Intestinal alkaline phosphatase detoxifies lipopolysaccharide and prevents inflammation in zebrafish in response to the gut microbiota [J]. Cell Host Microbe. 2007;2(6):371–382.
[16] Inami A, Kiyono H, Kurashima Y. ATP as a pathophysiologic mediator of bacteria-host crosstalk in the gastrointestinal tract [J]. Int J Mol Sci (2018) 19(8):2371.
[17] Bilski J, Mazur-Bialy A, et al. The role of intestinal alkaline phosphatase in inflammatory disorders of gastrointestinal tract [J]. Mediators Inflamm (2017) 2017:9074601.
[18] Vuerich M, Mukherjee S, Robson SC, Longhi MS. Control of gut inflammation by modulation of purinergic signaling [J]. Front Immunol (2020) 11:1882.
[19] Estaki M, DeCoffe D, Gibson DL. Interplay between intestinal alkaline phosphatase, diet, gut microbes and immunity [J]. World J Gastroenterol (2014) 20(42):15650–6.
[20] Goldberg R. F., Austen W. G., Jr., Zhang X., et al. Intestinal alkaline phosphatase is a gut mucosal defense factor maintained by enteral nutrition [J]. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(9):3551–3556.
[21] Malo M. S., Nasrin Alam S., Mostafa G., et al Intestinal alkaline phosphatase preserves the normal homeostasis of gut microbiota [J]. Gut. 2010;59(11):1476–1484.
[22] Malo M. S., Moaven O., Muhammad N., et al. Intestinal alkaline phosphatase promotes gut bacterial growth by reducing the concentration of luminal nucleotide triphosphates [J]. American Journal of Physiology—Gastrointestinal and Liver Physiology. 2014;306(10):G826–G838.
[23] Lallès J.-P. Luminal ATP: the missing link between intestinal alkaline phosphatase, the gut microbiota, and inflammation [J]? American Journal of Physiology—Gastrointestinal and Liver Physiology. 2014;306(10):G824–G825.
[24] Alam S. N., Yammine H., Ahmed R., et al. Oral supplementation with intestinal alkaline phosphatase: a novel therapy to treat Salmonella colitis [J]. Gastroenterology. 2012;142(5):p. S692.
[25] [8]Mizumori M, Ham M, Guth PH, Engel E, Kaunitz JD, Akiba Y. Intestinal alkaline phosphatase regulates protective surface microclimate pH in rat duodenum [J]. J Physiol. 2009;587(11):2537–2549.
[26] Santos GM, Ismael S, Morais J, Araújo JR, Faria A, Calhau C, Marques C. Intestinal Alkaline Phosphatase: A Review of This Enzyme Role in the Intestinal Barrier Function [J]. Microorganisms. 2022 Mar 30;10(4):746.
[27] Alam SN, Yammine H, et al. Intestinal alkaline phosphatase prevents antibiotic-induced susceptibility to enteric pathogens [J]. Proc Natl Acad Sci U S A. 2014;111(15):5678–5683.
[28] Martins RDS, Hulscher JBF, Timmer A, Kooi EMW, Poelstra K. Necrotizing enterocolitis: a potential protective role for intestinal alkaline phosphatase as lipopolysaccharide detoxifying enzyme [J]. Front Pediatr. 2024 Apr 30;12:1401090.
[29] Martins RDS, Kooi EMW, Poelstra K, Hulscher JBF. The role of intestinal alkaline phosphatase in the development of necrotizing enterocolitis [J]. Early Hum Dev. 2023 Aug;183:105797.
[30] Whitehouse JS, Riggle KM, Purpi DP, Mayer AN, Pritchard KA Jr, Oldham KT, Gourlay DM. The protective role of intestinal alkaline phosphatase in necrotizing enterocolitis [J]. J Surg Res. 2010 Sep;163(1):79-85.
[31] Pickkers, P., Heemskerk, S., et al. (2012). Alkaline phosphatase for treatment of sepsis-induced acute kidney injury: A prospective randomized double-blind placebo-controlled trial [J]. Critical Care, 16(1), R14.
[32] Kaliannan K, Hamarneh SR, et al. Intestinal alkaline phosphatase prevents metabolic syndrome in mice [J]. Proceedings of the National Academy of Sciences of the United States of America, 2013;110(17), 7003.
[33] Goldberg RF, Austen WG Jr, et al. Intestinal alkaline phosphatase is a gut mucosal defense factor maintained by enteral nutrition [J]. Proceedings of the National Academy of Sciences of the United States of America, 105(9), 3551.
Comments
Leave a Comment