Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
Next Article in Journal
Association of Methyl Donor Nutrients’ Dietary Intake and Cognitive Impairment in the Elderly Based on the Intestinal Microbiome
Previous Article in Journal
Exploring Determinants of Health-Related Quality of Life in Emerging Adults with Type 1 Diabetes Mellitus: A Cross-Sectional Analysis
Previous Article in Special Issue
Oleocanthal Protects C2C12 Myotubes against the Pro-Catabolic and Anti-Myogenic Action of Stimuli Able to Induce Muscle Wasting In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Therapeutic Potential of Bromelain: Applications, Benefits, and Mechanisms

1
Department of Medicine, Division of Cardiology, Wilf Family Cardiovascular Research Institute, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York, NY 10461, USA
2
Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University, 80131 Naples, Italy
3
Department of Advanced Biomedical Sciences, Federico II University Hospital, 80131 Naples, Italy
4
Vein Clinic, University of Brescia, 25100 Brescia, Italy
5
Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy
6
Casa di Cura “Montevergine”, 83013 Avellino, Italy
7
Department of Molecular Pharmacology, Einstein Institute for Aging Research, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2024, 16(13), 2060; https://doi.org/10.3390/nu16132060
Submission received: 11 April 2024 / Revised: 17 June 2024 / Accepted: 22 June 2024 / Published: 28 June 2024

Abstract

:
Bromelain is a mixture of proteolytic enzymes primarily extracted from the fruit and stem of the pineapple plant (Ananas comosus). It has a long history of traditional medicinal use in various cultures, particularly in Central and South America, where pineapple is native. This systematic review will delve into the history, structure, chemical properties, and medical indications of bromelain. Bromelain was first isolated and described in the late 19th century by researchers in Europe, who identified its proteolytic properties. Since then, bromelain has gained recognition in both traditional and modern medicine for its potential therapeutic effects.

1. Introduction

The history of bromelain dates back to the ancient civilizations of South America, where the pineapple plant (Ananas comosus) is native. Indigenous peoples in Central and South America, particularly in regions like the Amazon rainforest and the Caribbean, used various parts of the pineapple plant for medicinal purposes, including treating digestive issues, reducing inflammation, and healing wounds. The modern history of bromelain began in the late 19th century, when researchers started to explore the potential therapeutic properties of pineapple enzymes [1,2,3,4,5,6].
Originally, bromelain was exclusively derived from Hawaiian pineapple stems, but it is now also manufactured in Taiwan, Thailand, Brazil, and Puerto Rico. However, the variability in the commercially produced product and its diverse ingredients have hindered successful development. While pineapple bromelain has found commercial applications as a meat-tenderizing enzyme and a nutraceutical, efforts have been made to explore its potential for pharmaceutical use (Figure 1). Yet, the intricate nature of bromelain’s active components has posed some limitations on pharmaceutical research [7,8,9].
The history of bromelain spans centuries, from its traditional use by indigenous peoples to its modern industrial production and medical applications. Ongoing research continues to uncover new insights into its therapeutic potential and broaden its range of uses in various fields.

2. Chemical Properties

Bromelain is a complex mixture of proteolytic enzymes, including various proteases, such as stem bromelain, fruit bromelain, and ananain [10,11]. These enzymes belong to the cysteine protease family and exhibit different substrate specificities and optimal pH ranges [11,12,13,14]. The composition of bromelain can vary depending on factors such as the source of extraction and processing methods [15,16,17,18]. In more detail, bromelain activity occurs between pH 3 and 7. Beyond this value, it declines progressively, causing decreased absorption at higher pH values. Notably, several studies demonstrated that the proteolytic enzymes constituting bromelain mixture are absorbed from the gastrointestinal tract in an intact and functional form then available for intestinal absorption [19,20].
For the abovementioned reasons, bromelain is completely absorbed in any form and has no intestinal degradation problems.
Fruit Bromelain vs. Stem Bromelain
EC 3.4.22.33 (Fruit Bromelain) and EC 3.4.22.32 (Stem Bromelain) are both types of bromelain, a group of proteolytic enzymes derived from Ananas comosus that belong to the peptidase family C1 [21,22,23].
Data adapted from the BRaunschweig ENzyme Database (BRENDA), a collection of enzyme functional information available to the scientific community free of charge and maintained by the Leibniz Institute DSMZ as part of the Digital Diversity project, show that, while the two forms share similarities in their biochemical properties and functions, there are notable differences between them [24,25,26,27,28,29,30], as depicted in Table 1; they both belong to peptidase family C1 (papain family). Another cysteine endopeptidase with similar action on small molecule substrates, pinguinain (formerly EC 3.4.99.18), is obtained from the related plant, Bromelia pinguin, but pinguinain differs from fruit and stem bromelain in being inhibited by chicken cystatin [31,32].

3. Biological Effects

Bromelain exerts its therapeutic effects through a multifaceted mechanism of action, which contribute to the anti-inflammatory, analgesic, antiangiogenic, and antioxidant properties of bromelain, making it a promising candidate for the treatment of various inflammatory and oxidative stress-related disorders; its diverse biological effects depend on multiple mechanisms of action, including proteolytic activity, anti-inflammatory and immunomodulatory effects, fibrinolytic activity, antioxidant properties, and modulation of cell signaling pathways. These multifaceted mechanisms can be summarized as follows.

3.1. Proteolytic Activity

Protein digestion: bromelain primarily acts as a proteolytic enzyme; this property allows bromelain to aid in protein digestion, facilitating the breakdown and absorption of dietary proteins in the gastrointestinal tract.

3.2. Fibrinolytic Activity

Breakdown of fibrin, a key component of thrombi and scar tissue: by promoting fibrinolysis, bromelain may help prevent excessive blood clot formation and improve circulation [33,34,35,36,37,38,39].

3.3. Antioxidant Effects

Scavenging free radicals: bromelain exhibits antioxidant properties by scavenging free radicals and reactive oxygen species (ROS). By neutralizing oxidative stress, bromelain helps protect cells and tissues from damage caused by oxidative injury [40,41,42,43,44,45,46,47,48,49,50,51].

3.4. Immune Modulation

Enhancement of immune function: bromelain exhibits immunomodulatory properties by enhancing various aspects of immune function. It promotes the activity of immune cells such as macrophages, natural killer (NK) cells, and lymphocytes, thereby enhancing immune surveillance and defense mechanisms against pathogens. Bromelain also helps maintain a balanced cytokine profile by modulating the production of both proinflammatory and anti-inflammatory cytokines. This balance is crucial for proper immune function and inflammatory responses [25,52,53,54,55,56,57,58,59,60,61,62,63,64].
In vitro experiments have shown that bromelain has a significant impact on various immune cells. Specifically, bromelain can modulate surface adhesion molecules on T cells, macrophages, and NK cells, which are crucial for cell–cell interactions and for the immune response in general. Furthermore, it can induce the secretion of proinflammatory cytokines by peripheral blood mononuclear cells (PBMCs), playing instrumental roles in inflammation and immune regulation. Bromelain inhibits the T cell signal transduction pathways, particularly the Raf-1/extracellular-regulated-kinase- (ERK-) 2 pathway; this inhibition disrupts the signaling processes necessary for T cell activation [65]. Consequently, treatment with bromelain results in decreased activation of CD4+ T cells, a subset of T cells essential for orchestrating the immune response, and reduces the expression of CD25, a marker of T cell activation [66].
Bromelain has been reported to have analgesic and anti-inflammatory effects; these effects are thought to stem from its ability to influence pain mediators directly, such as bradykinin [34], which is involved in the sensation of pain and the inflammatory response [67]. By modulating these pathways and mediators, bromelain demonstrates potential therapeutic benefits for conditions characterized by inflammation and pain.

3.5. Regulation of Specific Cell Signaling Pathways

PI3K/Akt and MAPK pathways: bromelain influences various cell signaling pathways involved in cell proliferation, survival, and inflammation. It modulates the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) pathway and the mitogen-activated protein kinase (MAPK) pathway, thereby regulating cellular responses to extracellular stimuli [55,56,57,61,68,69,70,71,72,73,74,75,76,77,78,79,80].
Studies in Sprague–Dawley rats demonstrated that the beneficial effects of bromelain are partly due to its ability to phosphorylate Akt, which, in turn, leads to the phosphorylation of FOXO3A [72].
Bromelain was also shown to exert inhibitory effects against LPS-stimulated inflammatory responses in RAW264.7 macrophage cells: the inhibition of iNOS and COX-2 expression by bromelain attenuated the production of IL-6 and TNF-α; the beneficial effects of bromelain on LPS-induced inflammatory responses were mainly associated with decreased expression of proteins involved in the NF-κB and MAPKs signaling pathways [56].

3.6. Down-Regulation of Plasma Kininogen

Bromelain has been shown to down-regulate plasma kininogen levels. Kininogens are precursors to kinins, which are potent mediators of inflammation and vasodilation [81,82]. By reducing plasma kininogen levels, bromelain may inhibit the production of kinins, thereby attenuating inflammation and its associated symptoms such as pain and swelling [34,81,82,83,84,85,86,87].

3.7. Inhibition of Prostaglandin E2 Expression

Prostaglandin E2 (PGE2) is a key mediator of inflammation and pain. It is synthesized from arachidonic acid by the enzyme cyclooxygenase (COX). Bromelain has been reported to inhibit the expression of COX enzymes, particularly COX-2, which are responsible for the synthesis of prostaglandins from arachidonic acid, thereby reducing the production of PGE2 [34,52,54,56,59,68,69,70,73,88,89,90,91]. By inhibiting PGE2 synthesis, bromelain helps mitigate inflammation and alleviate pain.

3.8. Degradation of Advanced Glycation End Product Receptors

Advanced glycation end products (AGEs) are formed through nonenzymatic glycation and oxidation of proteins, lipids, and nucleic acids [61,92,93]. Accumulation of AGEs is associated with various pathological conditions, including diabetes, cardiovascular disease, and neurodegenerative disorders. Bromelain has been shown to degrade AGE receptors, such as the receptor for AGEs (RAGE), thereby reducing AGE-induced inflammation and tissue damage.

3.9. Regulation of Angiogenesis

Angiogenesis, the formation of new blood vessels from pre-existing ones, plays a crucial role in various physiological and pathological processes, including wound healing, tumor growth, and inflammation. Bromelain has been reported to regulate angiogenic biomarkers, including vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs) [59,72,94,95,96,97]. By modulating angiogenic factors, bromelain may influence vascular remodeling and tissue repair processes.
The main biological activities and applications of fruit bromelain and stem bromelain are summarized in Table 2.

4. Medical Indications

Numerous clinical trials have demonstrated the efficacy and safety of bromelain across various medical conditions, including osteoarthritis, sinusitis, surgical wounds, cardiovascular health, and digestive health [98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118]. However, larger-scale studies and further research are needed to confirm these findings and establish optimal dosing regimens and treatment protocols for different indications. Long-term safety data and potential drug interactions should be carefully evaluated in future studies. Currently, bromelain is considered to be nontoxic and without side effects. For instance, Castel and collaborators [119] demonstrated that the body can absorb a significant amount of bromelain, ~12 gm/day, without any major side effects. Other studies reported daily doses from 200 up to 2000 mg for prolonged periods of time and without concerns [28,120]. Notably, albeit bromelain has shown therapeutic benefit in doses as small as 160 mg/day, it is worth mentioning that the best results occur when starting at a dose of 750–1000 mg/day [18,28,120]. While bromelain is generally considered safe, some individuals may experience mild side effects, including gastrointestinal disturbances (nausea, diarrhea, and abdominal discomfort) and allergic reactions (including skin rashes, itching, and swelling of the lips, tongue, or throat). Bromelain may interact with certain medications, including some anticonvulsants, some antibiotics, and some anticoagulants (e.g., warfarin).
The main medical conditions in which bromelain has been studied for its potential therapeutic effects are outlined in the following sections.

4.1. Inflammation, Edema, and Swelling

Bromelain is believed to have anti-inflammatory properties, which may help reduce swelling, pain, and inflammation associated with conditions such as arthritis, sports injuries, and postoperative recovery. Indeed, bromelain has been shown to inhibit the production of various inflammatory mediators, including cytokines, prostaglandins, and leukotrienes. These molecules play crucial roles in the initiation and propagation of inflammation.
Bromelain exerts potent anti-inflammatory effects by modulating various inflammatory mediators, including cytokines, chemokines, and prostaglandins. It inhibits the production of proinflammatory cytokines such as interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6) [39,50,53,54,55,58,61,62,64,121,122,123,124].
Additionally, bromelain suppresses the nuclear factor-kappa B (NF-κB) signaling pathway, a key regulator of inflammation and immune responses; by inhibiting NF-κB activation, bromelain reduces the expression of inflammatory genes and attenuates the inflammatory cascade [56,57,125]. Bromelain’s proteolytic activity facilitates the breakdown of proteins involved in edema formation and swelling. By degrading extracellular matrix proteins and reducing the accumulation of fluid in tissues, bromelain may help alleviate inflammation-associated edema [113,126,127,128,129,130]. This property has led to the use of bromelain in various conditions characterized by edema and swelling, such as acute injuries, postoperative recovery, and inflammatory joint disorders like arthritis [131,132,133]. A recent clinical study has shown that the association of bromelain with vitamin C in postoperative bimalleolar surgery led to better outcomes, allowing a reduction in complications [134].
Notably, as mentioned above, bromelain has immunomodulatory effects that may influence the inflammatory response. It can modulate the activity of immune cells such as macrophages, lymphocytes, and dendritic cells, which play critical roles in initiating and regulating inflammation; actually, several studies suggest that bromelain may promote a shift towards anti-inflammatory immune responses by modulating the balance between proinflammatory and anti-inflammatory cytokines and chemokines [56,135,136,137,138,139,140]. All these beneficial actions have also suggested that bromelain could have a role in promoting tissue repair [106,141,142,143,144].

4.2. Digestive Health

Bromelain has been traditionally used to aid digestion and alleviate symptoms of indigestion, bloating, and heartburn. It is thought to assist in the breakdown of proteins in the digestive tract [18,51,145,146,147,148]. Bromelain has been shown to impact various aspects of digestion. Its proteolytic activity aids in the breakdown of dietary proteins into smaller peptides and amino acids. By hydrolyzing peptide bonds within protein molecules, bromelain facilitates their digestion and absorption in the gastrointestinal tract. This property of bromelain may be particularly beneficial for individuals with impaired protein digestion, such as those with pancreatic insufficiency or digestive enzyme deficiencies [147,149,150,151,152]. Through its proteolytic action, bromelain may enhance the bioavailability of nutrients derived from proteins. By breaking down protein complexes into simpler forms, bromelain may facilitate the absorption of essential amino acids and peptides across the intestinal mucosa. An improved protein digestion and nutrient absorption can contribute to overall nutritional status and support various physiological functions in the body. In addition, bromelain exhibits anti-inflammatory properties that may help alleviate digestive discomfort associated with inflammation in the gastrointestinal tract. By modulating inflammatory mediators and reducing mucosal inflammation, bromelain may mitigate symptoms such as bloating, gas, and abdominal pain. Numerous studies suggest that bromelain supplementation may be beneficial for individuals with inflammatory conditions of the digestive tract, including inflammatory bowel disease (IBD) and gastritis [10,25,62,141,153,154,155,156,157]. Bromelain may indirectly support the production and secretion of digestive enzymes by stimulating pancreatic and intestinal function. By promoting a favorable environment for enzymatic activity, bromelain may enhance overall digestive capacity [158]. Hence, bromelain’s anti-inflammatory effects may help protect pancreatic and intestinal tissues from damage, thereby preserving their functional integrity and ensuring optimal enzyme secretion.
Henceforth, Bromelain’s proteolytic activity, anti-inflammatory properties, and potential modulation of gut microbiota collectively contribute to its beneficial effects on digestive health. Whether consumed as part of fresh pineapple or in supplement form, bromelain may support various aspects of digestion, including protein digestion, nutrient absorption, relief of digestive discomfort, and maintenance of gut microbiota balance. However, further research is needed to elucidate the specific mechanisms underlying bromelain’s effects on digestive health and its optimal therapeutic applications.

4.3. Aging

The relationship between bromelain and aging represents an area of emerging interest, though direct studies specifically addressing this relationship are limited [159,160]. Cellular senescence is a state in which cells cease to divide but do not die, contributing to aging and the development of age-related diseases. Senescent cells exhibit a senescence-associated secretory phenotype (SASP), which includes the secretion of proinflammatory cytokines, chemokines, and proteases that can lead to tissue dysfunction and promote various diseases. Bromelain, a complex mixture of proteolytic enzymes derived from the pineapple plant, has been studied for its various biological activities, including anti-inflammatory, immunomodulatory, and potential anticancer effects. While direct research on bromelain’s impact on cellular senescence is not extensively documented, its known properties suggest several potential mechanisms through which it could influence senescence and the aging process, including anti-inflammatory effects, immunomodulatory properties, and proteolytic activity. Indeed, bromelain has been shown to modulate the inflammatory process, which is closely linked to the development and persistence of cellular senescence. By potentially reducing inflammation, bromelain could indirectly affect the senescence-associated secretory phenotype, thereby mitigating the adverse effects of senescent cells on tissue function [161,162,163,164]. Bromelain’s ability to modulate the immune response could also play a role in managing the effects of cellular senescence. By influencing immune cell activity, bromelain might help in clearing senescent cells, a process known as immunosurveillance, thus contributing to the maintenance of tissue homeostasis and reducing age-related pathologies. The proteolytic activity of bromelain could be relevant in the context of senescence by modulating the extracellular matrix and affecting the tissue microenvironment [165]. This activity might influence the behavior of senescent cells and their interaction with surrounding cells and matrix components [166]. Although the potential relationship between bromelain and cellular senescence presents an intriguing avenue for research, more targeted studies are needed to elucidate the mechanisms by which bromelain may influence senescence and aging. Understanding these mechanisms could open new therapeutic strategies for aging and senescence-associated diseases, leveraging bromelain’s bioactive properties.

4.4. Dermatology

Bromelain has found various applications in dermatology and cosmetics, leveraging its unique properties to address a range of skin concerns and enhance cosmetic procedures [167,168,169]. Bromelain’s proteolytic activity may contribute to wound healing and tissue repair processes by facilitating the removal of damaged tissue and promoting the proliferation of healthy cells [170,171]; by accelerating the resolution of inflammation and supporting the remodeling of injured tissues, bromelain may enhance the body’s ability to recover from injuries, surgical procedures, and other forms of tissue damage [96,106,108,171,172,173,174,175,176,177,178,179,180,181,182,183]. Bromelain is utilized to manage bruising and swelling following nonsurgical cosmetic procedures. Its application, either topically or orally, can lead to a reduction in the development of bruises and may also accelerate the healing process. This is particularly relevant following procedures that may lead to ecchymosis and edema, where bromelain’s anti-inflammatory properties can be beneficial. Bromelain acts as an exfoliating agent in cosmetic products. Its proteolytic activity enables it to gently remove dead skin cells, promoting skin renewal and improving texture. This enzymatic exfoliation is considered a gentler alternative to physical or chemical exfoliants, making it suitable for sensitive skin types. In the development of skin care products, such as cleansing washes and moisturizing lotions, bromelain is incorporated for its protease activity. This inclusion is based on its ability to aid in skin rejuvenation, enhance penetration of other active ingredients, and maintain skin hydration. Beyond cosmetic enhancements, bromelain’s properties have been explored for therapeutic applications in dermatology [168]. Its anti-inflammatory and wound-healing capabilities suggest potential benefits in treating various skin conditions, although more targeted research is needed to fully establish these therapeutic uses [184]. The use of bromelain, along with other botanical extracts like arnica, has been also indicated to minimize complications such as bruising and swelling following filler injections and other cosmetic interventions [185,186]; this application underscores bromelain’s role in post-procedure care, enhancing patient outcomes and satisfaction. Thus, as a botanical cosmeceutical, bromelain is recognized for its potential in improving skin health and appearance. Its incorporation into cosmetic products is supported by its stability and efficacy in various formulations, contributing to its growing popularity in the cosmetic industry.

4.5. Infectious Disorders

Some studies suggest that bromelain exhibits antibacterial properties [187,188,189], which may help prevent wound infections and promote a sterile wound environment conducive to healing. By inhibiting the growth of pathogenic bacteria and other microorganisms, bromelain may reduce the risk of wound complications and facilitate the healing process [95,178,189,190,191,192,193,194,195,196,197,198]. Bromelain has been recognized for its natural antimicrobial properties, which make it a candidate for treating infections of bacterial origin. Studies have highlighted its effectiveness against a variety of pathogens, suggesting its potential in managing infections, caries, and periodontal diseases among other conditions. The mechanism behind its antimicrobial activity may involve the disruption of bacterial cell walls or inhibition of bacterial adhesion and colonization. A specific case where bromelain has shown promise is in the treatment of Pityriasis lichenoides chronica, an infectious skin disease. Relatively recent observations indicate that bromelain treatment may lead to the complete resolution of this condition [199], showcasing its potential as a therapeutic agent in dermatological infectious disorders. Intriguingly, recent investigations have also proposed bromelain as a potential therapeutic strategy against COVID-19, caused by the SARS-CoV-2 virus [200,201]. Akhter and colleagues discovered that bromelain, when used alone at concentrations of 50 and 100 µg/mL, as well as in combination with acetylcysteine at concentrations of 50 and 100 µg/20 mg/mL, can disrupt the integrity of spike and envelope proteins of the SARS-CoV-2 virus [201]. Pretreatment with bromelain significantly hindered SARS-CoV-2 viral binding in VeroE6 cells, resulting in decreased viral infection and reduced SARS-CoV-2 viral RNA copies within the cells. Moreover, the combination of bromelain’s multifunctional enzymatic properties with acetylcysteine’s potent ability to break disulfide bonds led to inhibition of SARS-CoV-2 infectivity [201,202,203].
Ismail Celik and collaborators conducted a study utilizing molecular docking and molecular dynamics simulation techniques to explore the potential of bromelain in combating different variants of SARS-CoV-2 by targeting its interaction with angiotensin converting enzyme 2 (ACE2); the researchers discovered that bromelain demonstrated favorable binding affinity to various variants of the receptor-binding domain (RBD), which is crucial for the binding of the virus to ACE2 [204]. However, further studies are needed to validate these findings before any definitive conclusions can be drawn. The exploration into bromelain’s binding interactions with components of the virus suggests that it could be helpful in the management of COVID-19 as well as other bacterial-mediated diseases. This aspect is particularly relevant given the ongoing global health crisis and the need for effective treatments against the virus. Bromelain’s effects on the immune system are also pertinent to its role in infectious disorders. It has been shown to induce macrophage apoptosis and activation, which are crucial processes in the immune response to infections. By modulating the immune system, bromelain can help in treating immune-mediated conditions and potentially enhance the body’s defense against infections. Lastly, the development of bromelain-capped gold nanoparticles has been recommended as a novel drug delivery system for treating infectious diseases, including catheter-associated urinary tract infections. This innovative approach highlights the versatility of bromelain in medical applications and its potential to improve the efficacy of existing treatments.

4.6. Cancer

The relationship between bromelain and cancer has been a subject of increasing interest within the scientific community, particularly in exploring its potential as a therapeutic agent. In fact, there is emerging evidence suggesting anticancer properties of bromelain, with studies indicating potential benefits in inhibiting tumor growth, preventing metastasis, and enhancing the effectiveness of chemotherapy [17,18,51,205,206,207,208,209,210,211,212,213,214,215]. While research is ongoing and more evidence is needed, several mechanisms have been proposed to explain bromelain’s effects on cancer cells, including inhibition of cancer cell proliferation and induction of apoptosis, antiangiogenic effects, modulation of inflammatory pathways, enhancement of immune function, as well as by increasing the sensitivity of cancer cells to therapeutic interventions [94,141,195,197,198,199,200]. A novel approach involving the combination of bromelain and acetylcysteine has been recently discussed for its implications in cancer therapy [200]. This combination targets mucins in cancer cells, which are involved in the progression and metastasis of cancer. Mounting evidence suggests potential for bromelain + acetylcysteine in enhancing the efficacy of chemotherapy [76,216,217], although further studies are needed to fully understand its therapeutic potential and mechanisms of action. In vitro assays have demonstrated that bromelain can induce apoptosis in cancer cells, including breast cancer cells (specifically GI-101A cells) [218]. This phenomenon suggests that bromelain may contribute to the inhibition of cancer cell growth and potentially enhance the effectiveness of conventional cancer treatments. The cytotoxic effects of both unfractionated and fractionated bromelain on colorectal cancer cells have been investigated, alone or in combination with chemotherapeutic agents [214]; the findings indicate that bromelain treatment results in reduced cell survival in colorectal cancer cells in a dose-dependent manner, highlighting its potential as a complementary therapy in colorectal cancer treatment. Several studies have also explored the ACE2-inhibitory effects of bromelain in colon cancer cells [219]. Given the role of ACE2 in various physiological processes and its implications in cancer, bromelain’s inhibitory effects could offer a novel approach to targeting cancer cells [220], although the specific mechanisms and clinical relevance require further investigation.

5. Conclusions

Bromelain is a natural enzyme complex with diverse potential therapeutic effects, ranging from anti-inflammatory and digestive properties to immune modulation, aging, and wound healing. While research on bromelain continues to expand, further well-designed clinical trials are needed to elucidate its mechanisms of action, optimal dosing regimens, and efficacy in various medical conditions. Since its anti-inflammatory and antioxidant activities could be synergic with vitamin C, a possible association with this ingredient could be of interest. Furthermore, as illustrated above, anti-inflammatory and antioxidant activities can be achieved by combining 1000 mg bromelain with 500 mg of vitamin C. Healthcare providers should be aware of potential side effects and drug interactions associated with bromelain supplementation to ensure safe and appropriate use in clinical practice.

Author Contributions

Conceptualization, V.T. and G.S.; methodology, U.K., E.C. and P.M.; validation, M.V.M. and P.M.; data curation, U.K. and G.S.; writing—original draft preparation, U.K. and V.T.; writing—review and editing, G.S. All authors have read and agreed to the published version of the manuscript.

Funding

The Santulli’s Lab is currently supported in part by the National Institutes of Health (NIH): National Heart, Lung, and Blood Institute (NHLBI: R01-HL164772, R01-HL159062, R01-HL146691, T32-HL144456, T32-HL172255), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK: R01-DK123259, R01-DK033823), National Center for Advancing Translational Sciences (NCATS: UL1-TR002556-06, UM1-TR004400) to G.S., by the American Heart Association (AHA, 24IPA1268813), by the Diabetes Action Research and Education Foundation (to G.S.), and by the Monique Weill-Caulier and Irma T. Hirschl Trusts (to G.S.). Urna Kansakar is supported by the AHA (23POST1026190).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hunter, R.G.; Henry, G.W.; Civin, W.H. The action of papain and bromelain on the uterus. Am. J. Obstet. Gynecol. 1957, 73, 875–880. [Google Scholar] [CrossRef] [PubMed]
  2. Lewis, A.J. Papain, ficin and bromelain in the detection of incomplete Rhesus antibodies. Br. J. Haematol. 1957, 3, 332–339. [Google Scholar] [CrossRef] [PubMed]
  3. Simmons, C.A. The relief of pain in spasmodic dysmenorrhoea by bromelain. Lancet 1958, 2, 827–830. [Google Scholar] [CrossRef] [PubMed]
  4. Duncan, S.L.; Lawrie, J.H.; Maclennan, H.R. Bromelain and the cervix uteri. Lancet 1960, 2, 1420–1422. [Google Scholar] [CrossRef] [PubMed]
  5. Murachi, T.; Neurath, H. Fractionation and specificity studies on stem bromelain. J. Biol. Chem. 1960, 235, 99–107. [Google Scholar] [CrossRef] [PubMed]
  6. Youssef, A.F. The uterine isthmus and its sphincter mechanism: A clinical and radiographic study. III. The effect of bromelain on the uterine isthmus. Am. J. Obstet. Gynecol. 1960, 79, 1161–1168. [Google Scholar] [CrossRef] [PubMed]
  7. Ota, S.; Fu, T.H.; Hirohata, R. Studies on bromelain. II. Its activation and fractionation. J. Biochem. 1961, 49, 532–537. [Google Scholar] [CrossRef] [PubMed]
  8. El-Gharbawi, M.; Whitaker, J.R. Fractionation and Partial Characterization of the Proteolytic Enzymes of Stem Bromelain. Biochemistry 1963, 2, 476–481. [Google Scholar] [CrossRef] [PubMed]
  9. Inagami, T.; Murachi, T. Kinetic Studies of Bromelain Catalysis. Biochemistry 1963, 2, 1439–1444. [Google Scholar] [CrossRef] [PubMed]
  10. Hale, L.P.; Greer, P.K.; Trinh, C.T.; James, C.L. Proteinase activity and stability of natural bromelain preparations. Int. Immunopharmacol. 2005, 5, 783–793. [Google Scholar] [CrossRef]
  11. Azarkan, M.; Maquoi, E.; Delbrassine, F.; Herman, R.; M’Rabet, N.; Calvo Esposito, R.; Charlier, P.; Kerff, F. Structures of the free and inhibitors-bound forms of bromelain and ananain from Ananas comosus stem and in vitro study of their cytotoxicity. Sci. Rep. 2020, 10, 19570. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, C.H.; Hsia, C.C.; Hu, P.A.; Yeh, C.H.; Chen, C.T.; Peng, C.L.; Wang, C.H.; Lee, T.S. Bromelain Ameliorates Atherosclerosis by Activating the TFEB-Mediated Autophagy and Antioxidant Pathways. Antioxidants 2022, 12, 72. [Google Scholar] [CrossRef] [PubMed]
  13. Sharma, M.; Gupta, N.; Pandey, E. Implications of nasal delivery of bromelain on its pharmacokinetics, tissue distribution and pharmacodynamic profile-A preclinical study. PLoS ONE 2022, 17, e0277849. [Google Scholar] [CrossRef] [PubMed]
  14. Sorokin, A.V.; Goncharova, S.S.; Lavlinskaya, M.S.; Holyavka, M.G.; Faizullin, D.A.; Zuev, Y.F.; Kondratyev, M.S.; Artyukhov, V.G. Complexation of Bromelain, Ficin, and Papain with the Graft Copolymer of Carboxymethyl Cellulose Sodium Salt and N-Vinylimidazole Enhances Enzyme Proteolytic Activity. Int. J. Mol. Sci. 2023, 24, 11246. [Google Scholar] [CrossRef] [PubMed]
  15. Feng, X.; Hang, S.; Zhou, Y.; Liu, Q.; Yang, H. Bromelain Kinetics and Mechanism on Myofibril from Golden Pomfret (Trachinotus blochii). J. Food Sci. 2018, 83, 2148–2158. [Google Scholar] [CrossRef] [PubMed]
  16. Ningrum, A.; Wardani, D.W.; Vanidia, N.; Sarifudin, A.; Kumalasari, R.; Ekafitri, R.; Kristanti, D.; Setiaboma, W.; Munawaroh, H.S.H. In Silico Approach of Glycinin and Conglycinin Chains of Soybean By-Product (Okara) Using Papain and Bromelain. Molecules 2022, 27, 6855. [Google Scholar] [CrossRef] [PubMed]
  17. Rajan, P.K.; Dunna, N.R.; Venkatabalasubramanian, S. A comprehensive overview on the anti-inflammatory, antitumor, and ferroptosis functions of bromelain: An emerging cysteine protease. Expert. Opin. Biol. Ther. 2022, 22, 615–625. [Google Scholar] [CrossRef] [PubMed]
  18. Sharma, G.; Vimal, A. Bromelain: An Enzyme Expanding its Horizon from Food to Pharmaceutical Industry. Curr. Pharm. Biotechnol. 2023, 24, 1715–1726. [Google Scholar] [CrossRef] [PubMed]
  19. Maurer, H.R. Bromelain: Biochemistry, pharmacology and medical use. Cell. Mol. Life Sci. 2001, 58, 1234–1245. [Google Scholar] [CrossRef]
  20. Smyth, R.D.; Brennan, R.; Martin, G.J. Systemic biochemical changes following the oral administration of a proteolytic enzyme, bromelain. Arch. Int. Pharmacodyn. Ther. 1962, 136, 230–236. [Google Scholar]
  21. Yamada, F.; Takahashi, N.; Murachi, T. Purification and characterization of a proteinase from pineapple fruit, fruit bromelain FA2. J. Biochem. 1976, 79, 1223–1234. [Google Scholar] [CrossRef] [PubMed]
  22. Ota, S.; Muta, E.; Katahira, Y.; Okamoto, Y. Reinvestigation of fractionation and some properties of the proteolytically active components of stem and fruit bromelains. J. Biochem. 1985, 98, 219–228. [Google Scholar] [CrossRef] [PubMed]
  23. Rowan, A.D.; Buttle, D.J.; Barrett, A.J. The cysteine proteinases of the pineapple plant. Biochem. J. 1990, 266, 869–875. [Google Scholar]
  24. Ramli, A.N.; Aznan, T.N.; Illias, R.M. Bromelain: From production to commercialisation. J. Sci. Food Agric. 2017, 97, 1386–1395. [Google Scholar] [CrossRef]
  25. Zhou, Z.; Wang, L.; Feng, P.; Yin, L.; Wang, C.; Zhi, S.; Dong, J.; Wang, J.; Lin, Y.; Chen, D.; et al. Inhibition of Epithelial TNF-alpha Receptors by Purified Fruit Bromelain Ameliorates Intestinal Inflammation and Barrier Dysfunction in Colitis. Front. Immunol. 2017, 8, 1468. [Google Scholar] [CrossRef]
  26. Ramli, A.N.M.; Manas, N.H.A.; Hamid, A.A.A.; Hamid, H.A.; Illias, R.M. Comparative structural analysis of fruit and stem bromelain from Ananas comosus. Food Chem. 2018, 266, 183–191. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, L.; Li, W.; Liu, Y.; Zhi, W.; Han, J.; Wang, Y.; Ni, L. Green separation of bromelain in food sample with high retention of enzyme activity using recyclable aqueous two-phase system containing a new synthesized thermo-responsive copolymer and salt. Food Chem. 2019, 282, 48–57. [Google Scholar] [CrossRef] [PubMed]
  28. Varilla, C.; Marcone, M.; Paiva, L.; Baptista, J. Bromelain, a Group of Pineapple Proteolytic Complex Enzymes (Ananas comosus) and Their Possible Therapeutic and Clinical Effects. A Summary. Foods 2021, 10, 2249. [Google Scholar] [CrossRef]
  29. Razali, R.; Fahrudin, F.A.; Subbiah, V.K.; Takano, K.; Budiman, C. Heterologous Expression and Catalytic Properties of Codon-Optimized Small-Sized Bromelain from MD2 Pineapple. Molecules 2022, 27, 6031. [Google Scholar] [CrossRef]
  30. Yow, A.G.; Bostan, H.; Young, R.; Valacchi, G.; Gillitt, N.; Perkins-Veazie, P.; Xiang, Q.J.; Iorizzo, M. Identification of bromelain subfamily proteases encoded in the pineapple genome. Sci. Rep. 2023, 13, 11605. [Google Scholar] [CrossRef]
  31. Looby, C.I.; Eaton, W.D. Effects of Bromelia pinguin (Bromeliaceae) on soil ecosystem function and fungal diversity in the lowland forests of Costa Rica. BMC Ecol. 2014, 14, 12. [Google Scholar] [CrossRef] [PubMed]
  32. Meza-Espinoza, L.; de Los Angeles Vivar-Vera, M.; de Lourdes Garcia-Magana, M.; Sayago-Ayerdi, S.G.; Chacon-Lopez, A.; Becerrea-Verdin, E.M.; Montalvo-Gonzalez, E. Enzyme activity and partial characterization of proteases obtained from Bromelia karatas fruit and compared with Bromelia pinguin proteases. Food Sci. Biotechnol. 2018, 27, 509–517. [Google Scholar] [CrossRef] [PubMed]
  33. Azarkan, M.; Gonzalez, M.M.; Esposito, R.C.; Errasti, M.E. Stem Bromelain Proteolytic Machinery: Study of the Effects of its Components on Fibrin (ogen) and Blood Coagulation. Protein Pept. Lett. 2020, 27, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
  34. Kritis, P.; Karampela, I.; Kokoris, S.; Dalamaga, M. The combination of bromelain and curcumin as an immune-boosting nutraceutical in the prevention of severe COVID-19. Metabol. Open 2020, 8, 100066. [Google Scholar] [CrossRef] [PubMed]
  35. Hikisz, P.; Bernasinska-Slomczewska, J. Beneficial Properties of Bromelain. Nutrients 2021, 13, 4313. [Google Scholar] [CrossRef] [PubMed]
  36. Maher, H.M.; Almomen, A.; Alzoman, N.Z.; Shehata, S.M.; Alanazi, A.A. Development and validation of UPLC-MS/MS method for the simultaneous quantification of anaplastic lymphoma kinase inhibitors, alectinib, ceritinib, and crizotinib in Wistar rat plasma with application to bromelain-induced pharmacokinetic interaction. J. Pharm. Biomed. Anal. 2021, 204, 114276. [Google Scholar] [CrossRef] [PubMed]
  37. Rafiei-Asl, S.; Khadjeh, G.; Jalali, S.M.; Jamshidian, J.; Rezaie, A. Protective Effects of Bromelain against Cadmium-Induced Pulmonary Intoxication in Rats: A Histopathologic and Cytologic Study. Arch. Razi Inst. 2021, 76, 1427–1436. [Google Scholar] [CrossRef] [PubMed]
  38. Hu, P.A.; Wang, S.H.; Chen, C.H.; Guo, B.C.; Huang, J.W.; Lee, T.S. New Mechanisms of Bromelain in Alleviating Non-Alcoholic Fatty Liver Disease-Induced Deregulation of Blood Coagulation. Nutrients 2022, 14, 2329. [Google Scholar] [CrossRef] [PubMed]
  39. Pereira, I.C.; Satiro Vieira, E.E.; de Oliveira Torres, L.R.; Carneiro da Silva, F.C.; de Castro, E.S.J.M.; Torres-Leal, F.L. Bromelain supplementation and inflammatory markers: A systematic review of clinical trials. Clin. Nutr. ESPEN 2023, 55, 116–127. [Google Scholar] [CrossRef] [PubMed]
  40. Rasheedi, S.; Haq, S.K.; Khan, R.H. Guanidine hydrochloride denaturation of glycosylated and deglycosylated stem bromelain. Biochemistry 2003, 68, 1097–1100. [Google Scholar] [CrossRef]
  41. Sawano, Y.; Hatano, K.; Tanokura, M. Susceptibility of the interchain peptide of a bromelain inhibitor precursor to the target proteases bromelain, chymotrypsin, and trypsin. Biol. Chem. 2005, 386, 491–498. [Google Scholar] [CrossRef] [PubMed]
  42. Bakare, A.O.; Owoyele, B.V. Antinociceptive and neuroprotective effects of bromelain in chronic constriction injury-induced neuropathic pain in Wistar rats. Korean J. Pain. 2020, 33, 13–22. [Google Scholar] [CrossRef] [PubMed]
  43. El-Demerdash, F.M.; Baghdadi, H.H.; Ghanem, N.F.; Mhanna, A.B.A. Nephroprotective role of bromelain against oxidative injury induced by aluminium in rats. Environ. Toxicol. Pharmacol. 2020, 80, 103509. [Google Scholar] [CrossRef] [PubMed]
  44. Hu, P.A.; Chen, C.H.; Guo, B.C.; Kou, Y.R.; Lee, T.S. Bromelain Confers Protection against the Non-Alcoholic Fatty Liver Disease in Male C57bl/6 Mice. Nutrients 2020, 12, 1458. [Google Scholar] [CrossRef] [PubMed]
  45. Hu, R.; Chen, G.; Li, Y. Production and Characterization of Antioxidative Hydrolysates and Peptides from Corn Gluten Meal Using Papain, Ficin, and Bromelain. Molecules 2020, 25, 4091. [Google Scholar] [CrossRef] [PubMed]
  46. Jebur, A.B.; El-Demerdash, F.M.; Kang, W. Bromelain from Ananas comosus stem attenuates oxidative toxicity and testicular dysfunction caused by aluminum in rats. J. Trace Elem. Med. Biol. 2020, 62, 126631. [Google Scholar] [CrossRef] [PubMed]
  47. Lopez-Pedrouso, M.; Borrajo, P.; Pateiro, M.; Lorenzo, J.M.; Franco, D. Antioxidant activity and peptidomic analysis of porcine liver hydrolysates using alcalase, bromelain, flavourzyme and papain enzymes. Food Res. Int. 2020, 137, 109389. [Google Scholar] [CrossRef] [PubMed]
  48. Mekkawy, M.H.; Fahmy, H.A.; Nada, A.S.; Ali, O.S. Study of the Radiosensitizing and Radioprotective Efficacy of Bromelain (a Pineapple Extract): In Vitro and In Vivo. Integr. Cancer Ther. 2020, 19, 1534735420950468. [Google Scholar] [CrossRef]
  49. Mekkawy, M.H.; Fahmy, H.A.; Nada, A.S.; Ali, O.S. Radiosensitizing Effect of Bromelain Using Tumor Mice Model via Ki-67 and PARP-1 Inhibition. Integr. Cancer Ther. 2021, 20, 15347354211060369. [Google Scholar] [CrossRef]
  50. El-Demerdash, F.M.; Hussien, D.M.; Ghanem, N.F.; Al-Farga, A.M. Bromelain Modulates Liver Injury, Hematological, Molecular, and Biochemical Perturbations Induced by Aluminum via Oxidative Stress Inhibition. Biomed. Res. Int. 2022, 2022, 5342559. [Google Scholar] [CrossRef]
  51. Parasuraman, R.; Jayamurali, D.; Manoharan, N.; Govindarajulu, S.N. Effect of Bromelain on Chronic Unpredictable Stress-induced Behavioral, Biochemical, and Monoamine Changes in Wistar Albino Rat Model of Depression. Protein Pept. Lett. 2023, 30, 411–426. [Google Scholar] [CrossRef]
  52. Karlsen, M.; Hovden, A.O.; Vogelsang, P.; Tysnes, B.B.; Appel, S. Bromelain treatment leads to maturation of monocyte-derived dendritic cells but cannot replace PGE2 in a cocktail of IL-1beta, IL-6, TNF-alpha and PGE2. Scand. J. Immunol. 2011, 74, 135–143. [Google Scholar] [CrossRef] [PubMed]
  53. Bakare, A.O.; Owoyele, B.V. Bromelain reduced pro-inflammatory mediators as a common pathway that mediate antinociceptive and anti-anxiety effects in sciatic nerve ligated Wistar rats. Sci. Rep. 2021, 11, 289. [Google Scholar] [CrossRef]
  54. Brochard, S.; Pontin, J.; Bernay, B.; Boumediene, K.; Conrozier, T.; Bauge, C. The benefit of combining curcumin, bromelain and harpagophytum to reduce inflammation in osteoarthritic synovial cells. BMC Complement. Med. Ther. 2021, 21, 261. [Google Scholar] [CrossRef]
  55. Hong, J.H.; Kim, M.R.; Lee, B.N.; Oh, W.M.; Min, K.S.; Im, Y.G.; Hwang, Y.C. Anti-Inflammatory and Mineralization Effects of Bromelain on Lipopolysaccharide-Induced Inflammation of Human Dental Pulp Cells. Medicina 2021, 57, 591. [Google Scholar] [CrossRef] [PubMed]
  56. Insuan, O.; Janchai, P.; Thongchuai, B.; Chaiwongsa, R.; Khamchun, S.; Saoin, S.; Insuan, W.; Pothacharoen, P.; Apiwatanapiwat, W.; Boondaeng, A.; et al. Anti-Inflammatory Effect of Pineapple Rhizome Bromelain through Downregulation of the NF-kappaB- and MAPKs-Signaling Pathways in Lipopolysaccharide (LPS)-Stimulated RAW264.7 Cells. Curr. Issues Mol. Biol. 2021, 43, 93–106. [Google Scholar] [CrossRef]
  57. Pothacharoen, P.; Chaiwongsa, R.; Chanmee, T.; Insuan, O.; Wongwichai, T.; Janchai, P.; Vaithanomsat, P. Bromelain Extract Exerts Antiarthritic Effects via Chondroprotection and the Suppression of TNF-alpha-Induced NF-kappaB and MAPK Signaling. Plants 2021, 10, 2273. [Google Scholar] [CrossRef] [PubMed]
  58. Sehirli, A.O.; Sayiner, S.; Savtekin, G.; Velioglu-Ogunc, A. Protective effect of bromelain on corrosive burn in rats. Burns 2021, 47, 1352–1358. [Google Scholar] [CrossRef]
  59. Quarta, S.; Santarpino, G.; Carluccio, M.A.; Calabriso, N.; Scoditti, E.; Siculella, L.; Damiano, F.; Maffia, M.; Verri, T.; De Caterina, R.; et al. Analysis of the Anti-Inflammatory and Anti-Osteoarthritic Potential of Flonat Fast((R)), a Combination of Harpagophytum Procumbens DC. ex Meisn., Boswellia Serrata Roxb., Curcuma longa L., Bromelain and Escin (Aesculus hippocastanum), Evaluated in In Vitro Models of Inflammation Relevant to Osteoarthritis. Pharmaceuticals 2022, 15, 1263. [Google Scholar] [CrossRef]
  60. Sharma, M.; Chaudhary, D. In vitro and in vivo implications of rationally designed bromelain laden core-shell hybrid solid lipid nanoparticles for oral administration in thrombosis management. Nanomedicine 2022, 42, 102543. [Google Scholar] [CrossRef]
  61. Lu, H.C.; Ng, M.Y.; Liao, Y.W.; Maekawa, S.; Lin, T.; Yu, C.C. Bromelain inhibits the inflammation and senescence effect in diabetic periodontitis: A preliminary in vitro study. J. Dent. Sci. 2023, 18, 659–665. [Google Scholar] [CrossRef]
  62. Madkhali, J.Y.; Hussein, R.H.; Alnahdi, H.S. Therapeutic effect of bromelain and papain on intestinal injury induced by indomethacin in male rats. Int. J. Health Sci. 2023, 17, 23–30. [Google Scholar]
  63. Mousavi Maleki, M.S.; Ebrahimi Kiasari, R.; Seyed Mousavi, S.J.; Hashemi-Moghaddam, H.; Shabani, A.A.; Madanchi, H.; Sardari, S. Bromelain-loaded nanocomposites decrease inflammatory and cytotoxicity effects of gliadin on Caco-2 cells and peripheral blood mononuclear cells of celiac patients. Sci. Rep. 2023, 13, 21180. [Google Scholar] [CrossRef]
  64. Paksoy, T.; Ustaoglu, G.; Sehirli, A.O.; Unsal, R.B.K.; Sayiner, S.; Orhan, K.; Ayci, N.B.; Cetinel, S.; Aksoy, U.; Ogunc, A.V. Effect of bromelain on periodontal destruction and alveolar bone in rats with experimental periodontitis. Int. Immunopharmacol. 2023, 121, 110446. [Google Scholar] [CrossRef]
  65. Mynott, T.L.; Ladhams, A.; Scarmato, P.; Engwerda, C.R. Bromelain, from pineapple stems, proteolytically blocks activation of extracellular regulated kinase-2 in T cells. J. Immunol. 1999, 163, 2568–2575. [Google Scholar] [CrossRef]
  66. Secor, E.R., Jr.; Singh, A.; Guernsey, L.A.; McNamara, J.T.; Zhan, L.; Maulik, N.; Thrall, R.S. Bromelain treatment reduces CD25 expression on activated CD4+ T cells in vitro. Int. Immunopharmacol. 2009, 9, 340–346. [Google Scholar] [CrossRef]
  67. Gambardella, J.; Sorriento, D.; Bova, M.; Rusciano, M.; Loffredo, S.; Wang, X.; Petraroli, A.; Carucci, L.; Mormile, I.; Oliveti, M.; et al. Role of Endothelial G Protein-Coupled Receptor Kinase 2 in Angioedema. Hypertension 2020, 76, 1625–1636. [Google Scholar] [CrossRef]
  68. Hou, R.C.; Chen, Y.S.; Huang, J.R.; Jeng, K.C. Cross-linked bromelain inhibits lipopolysaccharide-induced cytokine production involving cellular signaling suppression in rats. J. Agric. Food Chem. 2006, 54, 2193–2198. [Google Scholar] [CrossRef]
  69. Kalra, N.; Bhui, K.; Roy, P.; Srivastava, S.; George, J.; Prasad, S.; Shukla, Y. Regulation of p53, nuclear factor kappaB and cyclooxygenase-2 expression by bromelain through targeting mitogen-activated protein kinase pathway in mouse skin. Toxicol. Appl. Pharmacol. 2008, 226, 30–37. [Google Scholar] [CrossRef]
  70. Bhui, K.; Prasad, S.; George, J.; Shukla, Y. Bromelain inhibits COX-2 expression by blocking the activation of MAPK regulated NF-kappa B against skin tumor-initiation triggering mitochondrial death pathway. Cancer Lett. 2009, 282, 167–176. [Google Scholar] [CrossRef]
  71. Bhui, K.; Tyagi, S.; Prakash, B.; Shukla, Y. Pineapple bromelain induces autophagy, facilitating apoptotic response in mammary carcinoma cells. Biofactors 2010, 36, 474–482. [Google Scholar] [CrossRef] [PubMed]
  72. Juhasz, B.; Thirunavukkarasu, M.; Pant, R.; Zhan, L.; Penumathsa, S.V.; Secor, E.R., Jr.; Srivastava, S.; Raychaudhuri, U.; Menon, V.P.; Otani, H.; et al. Bromelain induces cardioprotection against ischemia-reperfusion injury through Akt/FOXO pathway in rat myocardium. Am. J. Physiol. Heart Circ. Physiol. 2008, 294, H1365–H1370. [Google Scholar] [CrossRef] [PubMed]
  73. Bhui, K.; Tyagi, S.; Srivastava, A.K.; Singh, M.; Roy, P.; Singh, R.; Shukla, Y. Bromelain inhibits nuclear factor kappa-B translocation, driving human epidermoid carcinoma A431 and melanoma A375 cells through G(2)/M arrest to apoptosis. Mol. Carcinog. 2012, 51, 231–243. [Google Scholar] [CrossRef] [PubMed]
  74. Dave, S.; Kaur, N.J.; Nanduri, R.; Dkhar, H.K.; Kumar, A.; Gupta, P. Inhibition of adipogenesis and induction of apoptosis and lipolysis by stem bromelain in 3T3-L1 adipocytes. PLoS ONE 2012, 7, e30831. [Google Scholar] [CrossRef] [PubMed]
  75. Amini, A.; Ehteda, A.; Masoumi Moghaddam, S.; Akhter, J.; Pillai, K.; Morris, D.L. Cytotoxic effects of bromelain in human gastrointestinal carcinoma cell lines (MKN45, KATO-III, HT29-5F12, and HT29-5M21). Onco Targets Ther. 2013, 6, 403–409. [Google Scholar] [CrossRef] [PubMed]
  76. Amini, A.; Masoumi-Moghaddam, S.; Ehteda, A.; Morris, D.L. Bromelain and N-acetylcysteine inhibit proliferation and survival of gastrointestinal cancer cells in vitro: Significance of combination therapy. J. Exp. Clin. Cancer Res. 2014, 33, 92. [Google Scholar] [CrossRef] [PubMed]
  77. Arshad, Z.I.; Amid, A.; Yusof, F.; Jaswir, I.; Ahmad, K.; Loke, S.P. Bromelain: An overview of industrial application and purification strategies. Appl. Microbiol. Biotechnol. 2014, 98, 7283–7297. [Google Scholar] [CrossRef] [PubMed]
  78. Fouz, N.; Amid, A.; Hashim, Y.Z. Gene expression analysis in MCF-7 breast cancer cells treated with recombinant bromelain. Appl. Biochem. Biotechnol. 2014, 173, 1618–1639. [Google Scholar] [CrossRef] [PubMed]
  79. Romano, B.; Fasolino, I.; Pagano, E.; Capasso, R.; Pace, S.; De Rosa, G.; Milic, N.; Orlando, P.; Izzo, A.A.; Borrelli, F. The chemopreventive action of bromelain, from pineapple stem (Ananas comosus L.), on colon carcinogenesis is related to antiproliferative and proapoptotic effects. Mol. Nutr. Food Res. 2014, 58, 457–465. [Google Scholar] [CrossRef]
  80. Muller, A.; Barat, S.; Chen, X.; Bui, K.C.; Bozko, P.; Malek, N.P.; Plentz, R.R. Comparative study of antitumor effects of bromelain and papain in human cholangiocarcinoma cell lines. Int. J. Oncol. 2016, 48, 2025–2034. [Google Scholar] [CrossRef]
  81. Oh-ishi, S. Fluid phase activation of Hageman factor (factor XII) in citrated human plasma by bromelain: An application to the indirect enzymatic assay for Hageman factor. Thromb. Res. 1982, 27, 619–623. [Google Scholar] [CrossRef] [PubMed]
  82. Lotz-Winter, H. On the pharmacology of bromelain: An update with special regard to animal studies on dose-dependent effects. Planta Med. 1990, 56, 249–253. [Google Scholar] [CrossRef] [PubMed]
  83. Felton, G.E. Does kinin released by pineapple stem bromelain stimulate production of prostaglandin E1-like compounds? Hawaii. Med. J. 1977, 36, 39–47. [Google Scholar] [PubMed]
  84. Mineshita, S.; Nagai, Y. Hydrolysis of bradykinin by stem bromelain. Jpn. J. Pharmacol. 1977, 27, 170–172. [Google Scholar] [CrossRef] [PubMed]
  85. Kumakura, S.; Yamashita, M.; Tsurufuji, S. Effect of bromelain on kaolin-induced inflammation in rats. Eur. J. Pharmacol. 1988, 150, 295–301. [Google Scholar] [CrossRef]
  86. Bahde, R.; Palmes, D.; Minin, E.; Stratmann, U.; Diller, R.; Haier, J.; Spiegel, H.U. Bromelain ameliorates hepatic microcirculation after warm ischemia. J. Surg. Res. 2007, 139, 88–96. [Google Scholar] [CrossRef] [PubMed]
  87. Sufian, K.N.; Hira, T.; Nakamori, T.; Furuta, H.; Asano, K.; Hara, H. Soybean beta-conglycinin bromelain hydrolysate stimulates cholecystokinin secretion by enteroendocrine STC-1 cells to suppress the appetite of rats under meal-feeding conditions. Biosci. Biotechnol. Biochem. 2011, 75, 848–853. [Google Scholar] [CrossRef]
  88. Kasemsuk, T.; Saengpetch, N.; Sibmooh, N.; Unchern, S. Improved WOMAC score following 16-week treatment with bromelain for knee osteoarthritis. Clin. Rheumatol. 2016, 35, 2531–2540. [Google Scholar] [CrossRef]
  89. Huang, J.R.; Wu, C.C.; Hou, R.C.; Jeng, K.C. Bromelain inhibits lipopolysaccharide-induced cytokine production in human THP-1 monocytes via the removal of CD14. Immunol. Investig. 2008, 37, 263–277. [Google Scholar] [CrossRef]
  90. Hidayat, M.; Prahastuti, S.; Riany, D.U.; Soemardji, A.A.; Suliska, N.; Garmana, A.N.; Assiddiq, B.F.; Hasan, K. Kidney therapeutic potential of peptides derived from the bromelain hydrolysis of green peas protein. Iran. J. Basic. Med. Sci. 2019, 22, 1016–1025. [Google Scholar] [CrossRef]
  91. Mohamad, N.E.; Abu, N.; Yeap, S.K.; Alitheen, N.B. Bromelain Enhances the Anti-tumor Effects of Cisplatin on 4T1 Breast Tumor Model In Vivo. Integr. Cancer Ther. 2019, 18, 1534735419880258. [Google Scholar] [CrossRef]
  92. Foroncewicz, B.; Mucha, K.; Heidland, A.; Ciszek, M.; Imiela, J.; Helle, F.; Paczek, L. Modulation of serum levels of sRAGE by bromelain in patients with chronic kidney disease: A pilot study. Pol. Arch. Med. Wewn. 2012, 122, 514–516. [Google Scholar] [CrossRef]
  93. Gross, P.; Seelert, H.; Meiser, P.; Muller, R. Characterization of bromelain indicates a molar excess of inhibitor vs. enzyme molecules, a Jacalin-like lectin and Maillard reaction products. J. Pharm. Biomed. Anal. 2020, 181, 113075. [Google Scholar] [CrossRef] [PubMed]
  94. Shoba, E.; Lakra, R.; Syamala Kiran, M.; Korrapati, P.S. Fabrication of core-shell nanofibers for controlled delivery of bromelain and salvianolic acid B for skin regeneration in wound therapeutics. Biomed. Mater. 2017, 12, 035005. [Google Scholar] [CrossRef]
  95. Shoba, E.; Lakra, R.; Kiran, M.S.; Korrapati, P.S. 3 D nano bilayered spatially and functionally graded scaffold impregnated bromelain conjugated magnesium doped hydroxyapatite nanoparticle for periodontal regeneration. J. Mech. Behav. Biomed. Mater. 2020, 109, 103822. [Google Scholar] [CrossRef] [PubMed]
  96. Fathi, A.N.; Babaei, S.; Babaei, S.; Baazm, M.; Sakhaie, H.; Babaei, S. Effect of bromelain on mast cell numbers and degranulation in diabetic rat wound healing. J. Wound Care 2022, 31, S4–S11. [Google Scholar] [CrossRef]
  97. Weinzierl, A.; Harder, Y.; Schmauss, D.; Menger, M.D.; Laschke, M.W. Bromelain Protects Critically Perfused Musculocutaneous Flap Tissue from Necrosis. Biomedicines 2022, 10, 1449. [Google Scholar] [CrossRef] [PubMed]
  98. Bhuvan Chandra, R.; Selvarasu, K.; Krishnan, M. Comparison of Efficacy of Combination of Bromelain, Rutocide, and Trypsin With Serratiopeptidase on Postoperative Sequelae Following Mandibular Third Molar Surgery: A Randomized Clinical Trial. Cureus 2023, 15, e48633. [Google Scholar] [CrossRef]
  99. Zhao, Y.; Dai, X.; Sun, X.; Zhang, Z.; Gao, H.; Gao, R. Combination of Shengji ointment and bromelain in the treatment of exposed tendons in diabetic foot ulcers: Study protocol for a non-blind, randomized, positive control clinical trial. BMC Complement. Med. Ther. 2023, 23, 359. [Google Scholar] [CrossRef]
  100. Faramarzi, M.; Sadighi, M.; Shirmohamadi, A.; Kazemi, R.; Zohdi, M. Effectiveness of Bromelain in the control of postoperative pain after periodontal surgery: A crossover randomized clinical trial. J. Adv. Periodontol. Implant. Dent. 2023, 15, 22–27. [Google Scholar] [CrossRef]
  101. Vosahlo, J.; Salus, A.; Smolko, M.; Nemcova, B.; Nordmeyer, V.; Mikles, M.; Rau, S.M.; Erik Johansen, O. Oral enzyme combination with bromelain, trypsin and the flavonoid rutoside reduces systemic inflammation and pain when used pre- and post-operatively in elective total hip replacement: A randomized exploratory placebo-controlled trial. Ther. Adv. Musculoskelet. Dis. 2023, 15, 1759720X231186875. [Google Scholar] [CrossRef] [PubMed]
  102. Babazade, H.; Mirzaagha, A.; Konarizadeh, S. The effect of bromelain in periodontal surgery: A double-blind randomized placebo-controlled trial. BMC Oral. Health 2023, 23, 286. [Google Scholar] [CrossRef] [PubMed]
  103. Della Volpe, A.; De Luca, P.; De Lucia, A.; Martines, F.; Piroli, P.; D’Ascanio, L.; Camaioni, A.; La Mantia, I.; Di Stadio, A. Single-Center-Single-Blinded Clinical Trial to Evaluate the Efficacy of a Nutraceutical Containing Boswellia Serrata, Bromelain, Zinc, Magnesium, Honey, Tyndallized Lactobacillus Acidophilus and Casei to Fight Upper Respiratory Tract Infection and Otitis Media. Healthcare 2022, 10, 1526. [Google Scholar] [CrossRef] [PubMed]
  104. Desideri, I.; Lucidi, S.; Francolini, G.; Meattini, I.; Ciccone, L.P.; Salvestrini, V.; Valzano, M.; Morelli, I.; Angelini, L.; Scotti, V.; et al. Use of an alfa-lipoic, Methylsulfonylmethane, Boswellia serrata and Bromelain dietary supplement (OPERA(R)) for aromatase inhibitors-related arthralgia management (AIA): A prospective phase II trial (NCT04161833). Med. Oncol. 2022, 39, 113. [Google Scholar] [CrossRef] [PubMed]
  105. Gupta, A.A.; Kambala, R.; Bhola, N.; Jadhav, A. Comparative efficacy of bromelain and aceclofenac in limiting post-operative inflammatory sequelae in surgical removal of lower impacted third molar: A randomized controlled, triple blind clinical trial. J. Dent. Anesth. Pain. Med. 2022, 22, 29–37. [Google Scholar] [CrossRef] [PubMed]
  106. Shoham, Y.; Shapira, E.; Haik, J.; Harats, M.; Egozi, D.; Robinson, D.; Kogan, L.; Elkhatib, R.; Telek, G.; Shalom, A. Bromelain-based enzymatic debridement of chronic wounds: Results of a multicentre randomized controlled trial. Wound Repair. Regen. 2021, 29, 899–907. [Google Scholar] [CrossRef] [PubMed]
  107. Pekas, E.J.; Shin, J.; Headid, R.J.; Son, W.M.; Layec, G.; Yadav, S.K.; Scott, S.D.; Park, S.Y. Combined anthocyanins and bromelain supplement improves endothelial function and skeletal muscle oxygenation status in adults: A double-blind placebo-controlled randomised crossover clinical trial. Br. J. Nutr. 2021, 125, 161–171. [Google Scholar] [CrossRef]
  108. Soheilifar, S.; Bidgoli, M.; Hooshyarfard, A.; Shahbazi, A.; Vahdatinia, F.; Khoshkhooie, F. Effect of Oral Bromelain on Wound Healing, Pain, and Bleeding at Donor Site Following Free Gingival Grafting: A Clinical Trial. J. Dent. 2018, 15, 309–316. [Google Scholar]
  109. Jayachandran, S.; Khobre, P. Efficacy of Bromelain along with Trypsin, Rutoside Trihydrate Enzymes and Diclofenac Sodium Combination Therapy for the treatment of TMJ Osteoarthritis—A Randomised Clinical Trial. J. Clin. Diagn. Res. 2017, 11, ZC09–ZC11. [Google Scholar] [CrossRef]
  110. Tadikonda, A.; Pentapati, K.C.; Urala, A.S.; Acharya, S. Anti-plaque and anti-gingivitis effect of Papain, Bromelain, Miswak and Neem containing dentifrice: A randomized controlled trial. J. Clin. Exp. Dent. 2017, 9, e649–e653. [Google Scholar] [CrossRef]
  111. Chandanwale, A.; Langade, D.; Sonawane, D.; Gavai, P. A Randomized, Clinical Trial to Evaluate Efficacy and Tolerability of Trypsin:Chymotrypsin as Compared to Serratiopeptidase and Trypsin:Bromelain:Rutoside in Wound Management. Adv. Ther. 2017, 34, 180–198. [Google Scholar] [CrossRef] [PubMed]
  112. Ley, C.M.; Ni, Q.; Liao, X.; Gao, H.L.; Robinson, N. Bromelain and cardiovascular risk factors in diabetes: An exploratory randomized, placebo controlled, double blind clinical trial. Chin. J. Integr. Med. 2016, 22, 728–737. [Google Scholar] [CrossRef] [PubMed]
  113. Majid, O.W.; Al-Mashhadani, B.A. Perioperative bromelain reduces pain and swelling and improves quality of life measures after mandibular third molar surgery: A randomized, double-blind, placebo-controlled clinical trial. J. Oral. Maxillofac. Surg. 2014, 72, 1043–1048. [Google Scholar] [CrossRef] [PubMed]
  114. de la Barrera-Nunez, M.C.; Yanez-Vico, R.M.; Batista-Cruzado, A.; Heurtebise-Saavedra, J.M.; Castillo-de Oyague, R.; Torres-Lagares, D. Prospective double-blind clinical trial evaluating the effectiveness of Bromelain in the third molar extraction postoperative period. Med. Oral. Patol. Oral. Cir. Bucal 2014, 19, e157–e162. [Google Scholar] [CrossRef] [PubMed]
  115. Muller, S.; Marz, R.; Schmolz, M.; Drewelow, B.; Eschmann, K.; Meiser, P. Placebo-controlled randomized clinical trial on the immunomodulating activities of low- and high-dose bromelain after oral administration—New evidence on the antiinflammatory mode of action of bromelain. Phytother. Res. 2013, 27, 199–204. [Google Scholar] [CrossRef]
  116. Inchingolo, F.; Tatullo, M.; Marrelli, M.; Inchingolo, A.M.; Picciariello, V.; Inchingolo, A.D.; Dipalma, G.; Vermesan, D.; Cagiano, R. Clinical trial with bromelain in third molar exodontia. Eur. Rev. Med. Pharmacol. Sci. 2010, 14, 771–774. [Google Scholar] [PubMed]
  117. Howat, R.C.; Lewis, G.D. The effect of bromelain therapy on episiotomy wounds--a double blind controlled clinical trial. J. Obstet. Gynaecol. Br. Commonw. 1972, 79, 951–953. [Google Scholar] [CrossRef]
  118. Deplazes, B.C.; Hofmaenner, D.A.; Scheier, T.C.; Epprecht, J.; Mayer, M.; Schweizer, T.A.; Buehler, P.K.; Frey, P.M.; Brugger, S.D. Enzymatic debridement with bromelain and development of bacteremia in burn injuries: A retrospective cohort study. Burns 2024, 50, 405–412. [Google Scholar] [CrossRef]
  119. Castell, J.V.; Friedrich, G.; Kuhn, C.S.; Poppe, G.E. Intestinal absorption of undegraded proteins in men: Presence of bromelain in plasma after oral intake. Am. J. Physiol. 1997, 273, G139–G146. [Google Scholar] [CrossRef]
  120. Pavan, R.; Jain, S.; Shraddha; Kumar, A. Properties and therapeutic application of bromelain: A review. Biotechnol. Res. Int. 2012, 2012, 976203. [Google Scholar] [CrossRef]
  121. Sahu, M.; Sharma, A.K.; Sharma, G.; Kumar, A.; Nandave, M.; Babu, V. Facile synthesis of bromelain copper nanoparticles to improve the primordial therapeutic potential of copper against acute myocardial infarction in diabetic rats. Can. J. Physiol. Pharmacol. 2022, 100, 210–219. [Google Scholar] [CrossRef]
  122. Ferah Okkay, I.; Okkay, U.; Bayram, C.; Cicek, B.; Sezen, S.; Aydin, I.C.; Mendil, A.S.; Hacimuftuoglu, A. Bromelain protects against cisplatin-induced ocular toxicity through mitigating oxidative stress and inflammation. Drug Chem. Toxicol. 2023, 46, 69–76. [Google Scholar] [CrossRef]
  123. Vellini, M.; Desideri, D.; Milanese, A.; Omini, C.; Daffonchio, L.; Hernandez, A.; Brunelli, G. Possible involvement of eicosanoids in the pharmacological action of bromelain. Arzneimittelforschung 1986, 36, 110–112. [Google Scholar]
  124. Agostinis, C.; Zorzet, S.; De Leo, R.; Zauli, G.; De Seta, F.; Bulla, R. The combination of N-acetyl cysteine, alpha-lipoic acid, and bromelain shows high anti-inflammatory properties in novel in vivo and in vitro models of endometriosis. Mediat. Inflamm. 2015, 2015, 918089. [Google Scholar] [CrossRef] [PubMed]
  125. Didamoony, M.A.; Atwa, A.M.; Abd El-Haleim, E.A.; Ahmed, L.A. Bromelain ameliorates D-galactosamine-induced acute liver injury: Role of SIRT1/LKB1/AMPK, GSK3beta/Nrf2 and NF-kappaB p65/TNF-alpha/caspase-8, -9 signalling pathways. J. Pharm. Pharmacol. 2022, 74, 1765–1775. [Google Scholar] [CrossRef] [PubMed]
  126. Ordesi, P.; Pisoni, L.; Nannei, P.; Macchi, M.; Borloni, R.; Siervo, S. Therapeutic efficacy of bromelain in impacted third molar surgery: A randomized controlled clinical study. Quintessence Int. 2014, 45, 679–684. [Google Scholar] [CrossRef] [PubMed]
  127. Bormann, K.H.; Weber, K.; Kloppenburg, H.; Staude, P.; Koch, A.; Meiser, P.; Gellrich, N.C. Perioperative Bromelain Therapy after Wisdom Teeth Extraction—A Randomized, Placebo-Controlled, Double-Blinded, Three-Armed, Cross-Over Dose-Finding Study. Phytother. Res. 2016, 30, 2012–2019. [Google Scholar] [CrossRef]
  128. Singh, T.; More, V.; Fatima, U.; Karpe, T.; Aleem, M.A.; Prameela, J. Effect of proteolytic enzyme bromelain on pain and swelling after removal of third molars. J. Int. Soc. Prev. Community Dent. 2016, 6, S197–S204. [Google Scholar] [CrossRef]
  129. Ghensi, P.; Cucchi, A.; Creminelli, L.; Tomasi, C.; Zavan, B.; Maiorana, C. Effect of Oral Administration of Bromelain on Postoperative Discomfort After Third Molar Surgery. J. Craniofac Surg. 2017, 28, e191–e197. [Google Scholar] [CrossRef]
  130. Consorti, G.; Monarchi, G.; Paglianiti, M.; Betti, E.; Balercia, P. Reduction of Post-Surgical Facial Edema Following Bromelain and Coumarin Intake in Traumatology: A Prospective Study with 100 Patients. J. Clin. Med. 2024, 13, 922. [Google Scholar] [CrossRef]
  131. Liu, S.; Zhao, H.; Wang, Y.; Zhao, H.; Ma, C. Oral Bromelain for the Control of Facial Swelling, Trismus, and Pain After Mandibular Third Molar Surgery: A Systematic Review and Meta-Analysis. J. Oral. Maxillofac. Surg. 2019, 77, 1566–1574. [Google Scholar] [CrossRef] [PubMed]
  132. Knackstedt, R.; Gatherwright, J. Perioperative Homeopathic Arnica and Bromelain: Current Results and Future Directions. Ann. Plast. Surg. 2020, 84, e10–e15. [Google Scholar] [CrossRef]
  133. Parrini, S.; De Ambrosi, C.; Chisci, G. The role of oral bromelain on “bad outcome” in mandibular third molar surgery. A split-mouth comparative study. Ann. Ital. Chir. 2023, 94, 332–335. [Google Scholar] [PubMed]
  134. Meccariello, L.; Bello, A.I.; Bove, G.; Gagliardo, N.; Raffaele, D.; Matera, L. The ion resonance and bromelain-vitamin C vs bromelainvitamin C to prevent ankle complications in post-operative bimalleolar surgery. Med. Glas. 2024, 21, 236–243. [Google Scholar] [CrossRef]
  135. Engwerda, C.R.; Andrew, D.; Murphy, M.; Mynott, T.L. Bromelain activates murine macrophages and natural killer cells in vitro. Cell. Immunol. 2001, 210, 5–10. [Google Scholar] [CrossRef]
  136. Barth, H.; Guseo, A.; Klein, R. In vitro study on the immunological effect of bromelain and trypsin on mononuclear cells from humans. Eur. J. Med. Res. 2005, 10, 325–331. [Google Scholar] [PubMed]
  137. Wen, S.; Huang, T.H.; Li, G.Q.; Yamahara, J.; Roufogalis, B.D.; Li, Y. Bromelain improves decrease in defecation in postoperative rats: Modulation of colonic gene expression of inducible nitric oxide synthase. Life Sci. 2006, 78, 995–1002. [Google Scholar] [CrossRef]
  138. Mahajan, S.; Chandra, V.; Dave, S.; Nanduri, R.; Gupta, P. Stem bromelain-induced macrophage apoptosis and activation curtail Mycobacterium tuberculosis persistence. J. Infect. Dis. 2012, 206, 366–376. [Google Scholar] [CrossRef]
  139. Schulz, A.; Fuchs, P.C.; Oplaender, C.; Valdez, L.B.; Schiefer, J.L. Effect of Bromelain-Based Enzymatic Debridement on Skin Cells. J. Burn. Care Res. 2018, 39, 527–535. [Google Scholar] [CrossRef]
  140. Michelini, S.; Fiorentino, A.; Cardone, M. Melilotus, Rutin and Bromelain in primary and secondary lymphedema. Lymphology 2019, 52, 177–186. [Google Scholar] [CrossRef]
  141. Sahbaz, A.; Aynioglu, O.; Isik, H.; Ozmen, U.; Cengil, O.; Gun, B.D.; Gungorduk, K. Bromelain: A natural proteolytic for intra-abdominal adhesion prevention. Int. J. Surg. 2015, 14, 7–11. [Google Scholar] [CrossRef]
  142. Ataide, J.A.; Cefali, L.C.; Rebelo, M.A.; Spir, L.G.; Tambourgi, E.B.; Jozala, A.F.; Chaud, M.V.; Silveira, E.; Gu, X.; Gava Mazzola, P. Bromelain Loading and Release from a Hydrogel Formulated Using Alginate and Arabic Gum. Planta Med. 2017, 83, 870–876. [Google Scholar] [CrossRef]
  143. Tan, Y.; Li, P. Bromelain has significant clinical benefits after extraction of the third molar during chemotherapy in patients with hematologic tumor. Oncol. Lett. 2018, 15, 2962–2966. [Google Scholar] [CrossRef]
  144. Bayat, S.; Amiri, N.; Pishavar, E.; Kalalinia, F.; Movaffagh, J.; Hashemi, M. Bromelain-loaded chitosan nanofibers prepared by electrospinning method for burn wound healing in animal models. Life Sci. 2019, 229, 57–66. [Google Scholar] [CrossRef] [PubMed]
  145. Chandrasekaran, S.; Luna-Vital, D.; de Mejia, E.G. Identification and Comparison of Peptides from Chickpea Protein Hydrolysates Using Either Bromelain or Gastrointestinal Enzymes and Their Relationship with Markers of Type 2 Diabetes and Bitterness. Nutrients 2020, 12, 3843. [Google Scholar] [CrossRef]
  146. Chakraborty, A.J.; Mitra, S.; Tallei, T.E.; Tareq, A.M.; Nainu, F.; Cicia, D.; Dhama, K.; Emran, T.B.; Simal-Gandara, J.; Capasso, R. Bromelain a Potential Bioactive Compound: A Comprehensive Overview from a Pharmacological Perspective. Life 2021, 11, 317. [Google Scholar] [CrossRef]
  147. Mekkawy, A.H.; Pillai, K.; Suh, H.; Badar, S.; Akhter, J.; Kepenekian, V.; Ke, K.; Valle, S.J.; Morris, D.L. Bromelain and acetylcysteine (BromAc((R))) alone and in combination with gemcitabine inhibit subcutaneous deposits of pancreatic cancer after intraperitoneal injection. Am. J. Transl. Res. 2021, 13, 13524–13539. [Google Scholar]
  148. Ebrahimian, M.; Mahvelati, F.; Malaekeh-Nikouei, B.; Hashemi, E.; Oroojalian, F.; Hashemi, M. Bromelain Loaded Lipid-Polymer Hybrid Nanoparticles for Oral Delivery: Formulation and Characterization. Appl. Biochem. Biotechnol. 2022, 194, 3733–3748. [Google Scholar] [CrossRef]
  149. Bernkop-Schnurch, A.; Giovanelli, R.; Valenta, C. Peroral administration of enzymes: Strategies to improve the galenic of dosage forms for trypsin and bromelain. Drug Dev. Ind. Pharm. 2000, 26, 115–121. [Google Scholar] [CrossRef] [PubMed]
  150. Pillai, K.; Akhter, J.; Chua, T.C.; Morris, D.L. Anticancer property of bromelain with therapeutic potential in malignant peritoneal mesothelioma. Cancer Investig. 2013, 31, 241–250. [Google Scholar] [CrossRef] [PubMed]
  151. Higashi, T.; Kogo, T.; Sato, N.; Hirotsu, T.; Misumi, S.; Nakamura, H.; Iohara, D.; Onodera, R.; Motoyama, K.; Arima, H. Efficient Anticancer Drug Delivery for Pancreatic Cancer Treatment Utilizing Supramolecular Polyethylene-Glycosylated Bromelain. ACS Appl. Bio Mater. 2020, 3, 3005–3014. [Google Scholar] [CrossRef] [PubMed]
  152. Pillai, K.; Mekkawy, A.H.; Akhter, J.; Badar, S.; Dong, L.; Liu, A.I.; Morris, D.L. Enhancing the potency of chemotherapeutic agents by combination with bromelain and N-acetylcysteine—An in vitro study with pancreatic and hepatic cancer cells. Am. J. Transl. Res. 2020, 12, 7404–7419. [Google Scholar] [PubMed]
  153. Hale, L.P. Proteolytic activity and immunogenicity of oral bromelain within the gastrointestinal tract of mice. Int. Immunopharmacol. 2004, 4, 255–264. [Google Scholar] [CrossRef] [PubMed]
  154. Hale, L.P.; Greer, P.K.; Trinh, C.T.; Gottfried, M.R. Treatment with oral bromelain decreases colonic inflammation in the IL-10-deficient murine model of inflammatory bowel disease. Clin. Immunol. 2005, 116, 135–142. [Google Scholar] [CrossRef] [PubMed]
  155. Fitzhugh, D.J.; Shan, S.; Dewhirst, M.W.; Hale, L.P. Bromelain treatment decreases neutrophil migration to sites of inflammation. Clin. Immunol. 2008, 128, 66–74. [Google Scholar] [CrossRef] [PubMed]
  156. Onken, J.E.; Greer, P.K.; Calingaert, B.; Hale, L.P. Bromelain treatment decreases secretion of pro-inflammatory cytokines and chemokines by colon biopsies in vitro. Clin. Immunol. 2008, 126, 345–352. [Google Scholar] [CrossRef] [PubMed]
  157. Bottega, R.; Persico, I.; De Seta, F.; Romano, F.; Di Lorenzo, G. Anti-inflammatory properties of a proprietary bromelain extract (Bromeyal) after in vitro simulated gastrointestinal digestion. Int. J. Immunopathol. Pharmacol. 2021, 35, 20587384211034686. [Google Scholar] [CrossRef] [PubMed]
  158. Lee, S.Y.; Kang, J.H.; Lee, D.Y.; Jeong, J.W.; Kim, J.H.; Moon, S.S.; Hur, S.J. Methods for improving meat protein digestibility in older adults. J. Anim. Sci. Technol. 2023, 65, 32–56. [Google Scholar] [CrossRef] [PubMed]
  159. Ajagun-Ogunleye, M.O.; Ebuehi, O.A.T. Evaluation of the anti-aging and antioxidant action of Ananas sativa and Moringa oleifera in a fruit fly model organism. J. Food Biochem. 2020, 44, e13426. [Google Scholar] [CrossRef] [PubMed]
  160. Hammer, M.; Muuss, M.; Schickhardt, S.; Scheuerle, A.; Khoramnia, R.; Labuz, G.; Uhl, P.; Auffarth, G.U. Forward Light Scattering of the Vitreous Gel After Enzymatic Aging: An In Vitro Model to Study Vitreous Opacification. Investig. Ophthalmol. Vis. Sci. 2024, 65, 36. [Google Scholar] [CrossRef]
  161. Abbasi Habashi, S.; Sabouni, F.; Moghimi, A.; Ansari Majd, S. Modulation of Lipopolysaccharide Stimulated Nuclear Factor kappa B Mediated iNOS/NO Production by Bromelain in Rat Primary Microglial Cells. Iran. Biomed. J. 2016, 20, 33–40. [Google Scholar] [CrossRef] [PubMed]
  162. Fujiwara, M.; Kariyone, A. Lipopolysaccharide-induced autoantibody response. II. Age-related change in plaque-forming cell response to bromelain-treated syngeneic erythrocytes. Int. Arch. Allergy Appl. Immunol. 1981, 66, 161–172. [Google Scholar] [CrossRef] [PubMed]
  163. Zhao, K.S.; Wang, Y.F.; Gueret, R.; Weksler, M.E. Dysregulation of the humoral immune response in old mice. Int. Immunol. 1995, 7, 929–934. [Google Scholar] [CrossRef] [PubMed]
  164. Bovbjerg, D.H.; Kim, Y.T.; Schwab, R.; Schmitt, K.; DeBlasio, T.; Weksler, M.E. “Cross-wiring” of the immune response in old mice: Increased autoantibody response despite reduced antibody response to nominal antigen. Cell. Immunol. 1991, 135, 519–525. [Google Scholar] [CrossRef] [PubMed]
  165. Gallego, M.; Grau, R.; Talens, P. Behaviour of texture-modified meats using proteolytic enzymes during gastrointestinal digestion simulating elderly alterations. Meat Sci. 2023, 206, 109341. [Google Scholar] [CrossRef] [PubMed]
  166. Zhai, Y.; Cui, Y.; Song, M.; Vainstein, A.; Chen, S.; Ma, H. Papain-Like Cysteine Protease Gene Family in Fig (Ficus carica L.): Genome-Wide Analysis and Expression Patterns. Front. Plant Sci. 2021, 12, 681801. [Google Scholar] [CrossRef] [PubMed]
  167. Baumann, L.S. Less-known botanical cosmeceuticals. Dermatol. Ther. 2007, 20, 330–342. [Google Scholar] [CrossRef] [PubMed]
  168. Harats, M.; Haik, J.; Cleary, M.; Vashurin, I.; Aviv, U.; Kornhaber, R. A Retrospective Review of an Off-label Bromelain-based Selective Enzymatic Debridement (Nexobrid(R)) in the Treatment of Deep, Partial, and Full Thickness Burns and Hard to Heal Wounds. Isr. Med. Assoc. J. 2020, 22, 83–88. [Google Scholar]
  169. Wang, S.; Li, J.; Ma, Z.; Sun, L.; Hou, L.; Huang, Y.; Zhang, Y.; Guo, B.; Yang, F. A Sequential Therapeutic Hydrogel With Injectability and Antibacterial Activity for Deep Burn Wounds’ Cleaning and Healing. Front. Bioeng. Biotechnol. 2021, 9, 794769. [Google Scholar] [CrossRef]
  170. Singer, A.J.; Toussaint, J.; Chung, W.T.; McClain, S.A.; Clark, R.A.F.; Asculai, E.; Geblinger, D.; Rosenberg, L. Development of a contaminated ischemic porcine wound model and the evaluation of bromelain based enzymatic debridement. Burns 2018, 44, 896–904. [Google Scholar] [CrossRef]
  171. Snyder, R.J.; Singer, A.J.; Dove, C.R.; Heisler, S.; Petusevsky, H.; James, G.; deLancey Pulcini, E.; Yaakov, A.B.; Rosenberg, L.; Grant, E.; et al. An open-label, proof-of-concept study assessing the effects of bromelain-based enzymatic debridement on biofilm and microbial loads in patients with venous leg ulcers and diabetic foot ulcers. Wounds 2023, 35, E414–E419. [Google Scholar] [CrossRef]
  172. Ataide, J.A.; de Carvalho, N.M.; Rebelo, M.A.; Chaud, M.V.; Grotto, D.; Gerenutti, M.; Rai, M.; Mazzola, P.G.; Jozala, A.F. Bacterial Nanocellulose Loaded with Bromelain: Assessment of Antimicrobial, Antioxidant and Physical-Chemical Properties. Sci. Rep. 2017, 7, 18031. [Google Scholar] [CrossRef] [PubMed]
  173. Shoham, Y.; Krieger, Y.; Tamir, E.; Silberstein, E.; Bogdanov-Berezovsky, A.; Haik, J.; Rosenberg, L. Bromelain-based enzymatic debridement of chronic wounds: A preliminary report. Int. Wound J. 2018, 15, 769–775. [Google Scholar] [CrossRef]
  174. Ho, W.; Jones, C.D.; Widdowson, D.; Bahia, H. Bromelain-based enzymatic debridement of e-cigarette burns: A single unit experience. J. Wound Care 2019, 28, 758–761. [Google Scholar] [CrossRef]
  175. Fathi, A.N.; Sakhaie, M.H.; Babaei, S.; Babaei, S.; Slimabad, F.; Babaei, S. Use of bromelain in cutaneous wound healing in streptozocin-induced diabetic rats: An experimental model. J. Wound Care 2020, 29, 488–495. [Google Scholar] [CrossRef] [PubMed]
  176. Hirche, C.; Kreken Almeland, S.; Dheansa, B.; Fuchs, P.; Governa, M.; Hoeksema, H.; Korzeniowski, T.; Lumenta, D.B.; Marinescu, S.; Martinez-Mendez, J.R.; et al. Eschar removal by bromelain based enzymatic debridement (Nexobrid(R)) in burns: European consensus guidelines update. Burns 2020, 46, 782–796. [Google Scholar] [CrossRef]
  177. Ataide, J.A.; Cefali, L.C.; Figueiredo, M.C.; Braga, L.E.O.; Ruiz, A.; Foglio, M.A.; Oliveira-Nascimento, L.; Mazzola, P.G. In vitro performance of free and encapsulated bromelain. Sci. Rep. 2021, 11, 10195. [Google Scholar] [CrossRef]
  178. Hasannasab, M.; Nourmohammadi, J.; Dehghan, M.M.; Ghaee, A. Immobilization of bromelain and ZnO nanoparticles on silk fibroin nanofibers as an antibacterial and anti-inflammatory burn dressing. Int. J. Pharm. 2021, 610, 121227. [Google Scholar] [CrossRef]
  179. Shoham, Y.; Sabbag, I.; Singer, A.J. Development of a porcine hard-to-heal wound model: Evaluation of a bromelain-based enzymatic debriding agent. J. Wound Care 2021, 30, VIi–VIx. [Google Scholar] [CrossRef] [PubMed]
  180. Sharaf, A.; Muthayya, P. Microbial profile of burn wounds managed with enzymatic debridement using bromelain-based agent, NexoBrid(R). Burns 2022, 48, 1618–1625. [Google Scholar] [CrossRef]
  181. Esen, E.; Bayar Muluk, N.; Vejselova Sezer, C.; Kutlu, H.M.; Cingi, C. Bromelain: A candidate to enhance wound healing after endonasal surgeries. Eur. Rev. Med. Pharmacol. Sci. 2023, 27, 33–38. [Google Scholar] [CrossRef] [PubMed]
  182. Shoham, Y.; Gasteratos, K.; Singer, A.J.; Krieger, Y.; Silberstein, E.; Goverman, J. Bromelain-based enzymatic burn debridement: A systematic review of clinical studies on patient safety, efficacy and long-term outcomes. Int. Wound J. 2023, 20, 4364–4383. [Google Scholar] [CrossRef] [PubMed]
  183. Singer, A.J.; Goradia, E.N.; Grandfield, S.; Zhang, N.; Shah, K.; McClain, S.A.; Sandoval, S.; Shoham, Y. A Comparison of Topical Agents for Eschar Removal in a Porcine Model: Bromelain-enriched vs Traditional Collagenase Agents. J. Burn. Care Res. 2023, 44, 408–413. [Google Scholar] [CrossRef] [PubMed]
  184. Baumann, L. Botanical ingredients in cosmeceuticals. J. Drugs Dermatol. 2007, 6, 1084–1088. [Google Scholar] [PubMed]
  185. Sherban, A.; Wang, J.V.; Geronemus, R.G. Growing role for arnica in cosmetic dermatology: Lose the bruise. J. Cosmet. Dermatol. 2021, 20, 2062–2068. [Google Scholar] [CrossRef] [PubMed]
  186. Hamman, M.S.; Goldman, M.P. Minimizing bruising following fillers and other cosmetic injectables. J. Clin. Aesthet. Dermatol. 2013, 6, 16–18. [Google Scholar] [PubMed]
  187. Abbas, S.; Shanbhag, T.; Kothare, A. Applications of bromelain from pineapple waste towards acne. Saudi J. Biol. Sci. 2021, 28, 1001–1009. [Google Scholar] [CrossRef] [PubMed]
  188. Jancic, U.; Gorgieva, S. Bromelain and Nisin: The Natural Antimicrobials with High Potential in Biomedicine. Pharmaceutics 2021, 14, 76. [Google Scholar] [CrossRef] [PubMed]
  189. Chandwani, N.D.; Maurya, N.; Nikhade, P.; Chandwani, J. Comparative evaluation of antimicrobial efficacy of calcium hydroxide, triple antibiotic paste and bromelain against Enterococcus faecalis: An In Vitro study. J. Conserv. Dent. 2022, 25, 63–67. [Google Scholar] [CrossRef]
  190. Praveen, N.C.; Rajesh, A.; Madan, M.; Chaurasia, V.R.; Hiremath, N.V.; Sharma, A.M. In vitro Evaluation of Antibacterial Efficacy of Pineapple Extract (Bromelain) on Periodontal Pathogens. J. Int. Oral. Health 2014, 6, 96–98. [Google Scholar]
  191. dos Anjos, M.M.; da Silva, A.A.; de Pascoli, I.C.; Mikcha, J.M.; Machinski, M., Jr.; Peralta, R.M.; de Abreu Filho, B.A. Antibacterial activity of papain and bromelain on Alicyclobacillus spp. Int. J. Food Microbiol. 2016, 216, 121–126. [Google Scholar] [CrossRef] [PubMed]
  192. Ghanbari, R.; Ebrahimpour, A. Separation and identification of bromelain-generated antibacterial peptides from Actinopyga lecanora. Food Sci. Biotechnol. 2018, 27, 591–598. [Google Scholar] [CrossRef] [PubMed]
  193. Chen, X.; Wang, X.; Wang, S.; Zhang, X.; Yu, J.; Wang, C. Mussel-inspired polydopamine-assisted bromelain immobilization onto electrospun fibrous membrane for potential application as wound dressing. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 110, 110624. [Google Scholar] [CrossRef]
  194. Avalos-Flores, E.; Lopez-Castillo, L.M.; Wielsch, N.; Hupfer, Y.; Winkler, R.; Magana-Ortiz, D. Protein extract of Bromelia karatas L. rich in cysteine proteases (ananain- and bromelain-like) has antibacterial activity against foodborne pathogens Listeria monocytogenes and Salmonella Typhimurium. Folia Microbiol. 2022, 67, 1–13. [Google Scholar] [CrossRef] [PubMed]
  195. Aldoski, M.R.N.; Selivany, B.J.; Sulaiman, T. Bromelain-based endodontic irrigant: Preparation, properties, and biocompatibility: An in-vitro study. Aust. Endod. J. 2023, 49 (Suppl. S1), 146–155. [Google Scholar] [CrossRef] [PubMed]
  196. Saghafi, Y.; Baharifar, H.; Najmoddin, N.; Asefnejad, A.; Maleki, H.; Sajjadi-Jazi, S.M.; Bonkdar, A.; Shams, F.; Khoshnevisan, K. Bromelain- and Silver Nanoparticle-Loaded Polycaprolactone/Chitosan Nanofibrous Dressings for Skin Wound Healing. Gels 2023, 9, 672. [Google Scholar] [CrossRef] [PubMed]
  197. Atiyeh, B.; Makkawi, K.; Beaineh, P. Burn Wounds and Enzymatic Debridement (Ed)—Past, Present, and Future. J. Burn. Care Res. 2024; irae059, (in press). [Google Scholar] [CrossRef]
  198. Jancic, U.; Trcek, J.; Verestiuc, L.; Vukomanovic, M.; Gorgieva, S. Bacterial nanocellulose loaded with bromelain and nisin as a promising bioactive material for wound debridement. Int. J. Biol. Macromol. 2024, 266, 131329. [Google Scholar] [CrossRef] [PubMed]
  199. Massimiliano, R.; Pietro, R.; Paolo, S.; Sara, P.; Michele, F. Role of bromelain in the treatment of patients with pityriasis lichenoides chronica. J. Dermatol. Treat. 2007, 18, 219–222. [Google Scholar] [CrossRef] [PubMed]
  200. Sagar, S.; Rathinavel, A.K.; Lutz, W.E.; Struble, L.R.; Khurana, S.; Schnaubelt, A.T.; Mishra, N.K.; Guda, C.; Palermo, N.Y.; Broadhurst, M.J.; et al. Bromelain inhibits SARS-CoV-2 infection via targeting ACE-2, TMPRSS2, and spike protein. Clin. Transl. Med. 2021, 11, e281. [Google Scholar] [CrossRef]
  201. Akhter, J.; Queromes, G.; Pillai, K.; Kepenekian, V.; Badar, S.; Mekkawy, A.H.; Frobert, E.; Valle, S.J.; Morris, D.L. The Combination of Bromelain and Acetylcysteine (BromAc) Synergistically Inactivates SARS-CoV-2. Viruses 2021, 13, 425. [Google Scholar] [CrossRef]
  202. Ferreira, G.M.; Clarindo, F.A.; Ribeiro, A.L.; Gomes-de-Pontes, L.; de Carvalho, L.D.; Martins-Filho, O.A.; da Fonseca, F.G.; Teixeira, M.M.; Sabino, A.P.; Eapen, M.S.; et al. Taming the SARS-CoV-2-mediated proinflammatory response with BromAc((R)). Front. Immunol. 2023, 14, 1308477. [Google Scholar] [CrossRef] [PubMed]
  203. Yang, J.; Glenn, D.; Lodh, S.; Valle, S.; Morris, D.L. Long-Term Treatment of Unresectable Pseudomyxoma Peritonei with Multiple Treatments of Intratumoural Bromelain and Acetylcysteine (BromAc((R))): A Case Report. Case Rep. Oncol. 2023, 16, 1551–1556. [Google Scholar] [CrossRef] [PubMed]
  204. Tallei, T.E.; Fatimawali; Yelnetty, A.; Idroes, R.; Kusumawaty, D.; Emran, T.B.; Yesiloglu, T.Z.; Sippl, W.; Mahmud, S.; Alqahtani, T.; et al. An Analysis Based on Molecular Docking and Molecular Dynamics Simulation Study of Bromelain as Anti-SARS-CoV-2 Variants. Front. Pharmacol. 2021, 12, 717757. [Google Scholar] [CrossRef] [PubMed]
  205. Park, S.; Oh, J.; Kim, M.; Jin, E.J. Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim. Cells Syst. 2018, 22, 334–340. [Google Scholar] [CrossRef] [PubMed]
  206. Chang, T.C.; Wei, P.L.; Makondi, P.T.; Chen, W.T.; Huang, C.Y.; Chang, Y.J. Bromelain inhibits the ability of colorectal cancer cells to proliferate via activation of ROS production and autophagy. PLoS ONE 2019, 14, e0210274. [Google Scholar] [CrossRef] [PubMed]
  207. Taskin, A.; Tarakcioglu, M.; Ulusal, H.; Orkmez, M.; Taysi, S. Idarubicin-bromelain combination sensitizes cancer cells to conventional chemotherapy. Iran. J. Basic. Med. Sci. 2019, 22, 1172–1178. [Google Scholar] [CrossRef]
  208. Amini Chermahini, F.; Raeisi, E.; Aazami, M.H.; Mirzaei, A.; Heidarian, E.; Lemoigne, Y. Does Bromelain-Cisplatin Combination Afford In-Vitro Synergistic Anticancer Effects on Human Prostatic Carcinoma Cell Line, PC3? Galen. Med. J. 2020, 9, e1749. [Google Scholar] [CrossRef]
  209. Pezzani, R.; Jimenez-Garcia, M.; Capo, X.; Sonmez Gurer, E.; Sharopov, F.; Rachel, T.Y.L.; Ntieche Woutouoba, D.; Rescigno, A.; Peddio, S.; Zucca, P.; et al. Anticancer properties of bromelain: State-of-the-art and recent trends. Front. Oncol. 2022, 12, 1068778. [Google Scholar] [CrossRef] [PubMed]
  210. Ricardo, P.C.; Serudo, R.L.; Talu, S.; Lamarao, C.V.; da Fonseca Filho, H.D.; de Araujo Bezerra, J.; Sanches, E.A.; Campelo, P.H. Encapsulation of Bromelain in Combined Sodium Alginate and Amino Acid Carriers: Experimental Design of Simplex-Centroid Mixtures for Digestibility Evaluation. Molecules 2022, 27, 6364. [Google Scholar] [CrossRef]
  211. Zhu, X.; Wang, M.; Wang, H.; Ding, Y.; Liu, Y.; Fu, Z.; Lin, D.; Lu, C.; Tu, X. Multifunctional Hollow MnO(2) @Porphyrin@Bromelain Nanoplatform for Enhanced Photodynamic Therapy. Small 2022, 18, e2204951. [Google Scholar] [CrossRef]
  212. Kumar, V.; Mangla, B.; Javed, S.; Ahsan, W.; Kumar, P.; Garg, V.; Dureja, H. Bromelain: A review of its mechanisms, pharmacological effects and potential applications. Food Funct. 2023, 14, 8101–8128. [Google Scholar] [CrossRef]
  213. Skalkos, E.; Chen, K.L.; Wijayawardana, R.; Morris, D.L. Effective Use of Bromelain and Acetylcysteine (BromAc((R))) for Treatment of Perigastric Pseudomyxoma Peritonei: A Case Report. Anticancer. Res. 2023, 43, 4735–4738. [Google Scholar] [CrossRef] [PubMed]
  214. Tsai, K.Y.; Wei, P.L.; Azarkan, M.; M’Rabet, N.; Makondi, P.T.; Chen, H.A.; Huang, C.Y.; Chang, Y.J. Cytotoxic properties of unfractionated and fractionated bromelain alone or in combination with chemotherapeutic agents in colorectal cancer cells. PLoS ONE 2023, 18, e0285970. [Google Scholar] [CrossRef]
  215. Wen, H.K.; Valle, S.J.; Morris, D.L. Bromelain and acetylcysteine (BromAc((R))): A novel approach to the treatment of mucinous tumours. Am. J. Cancer Res. 2023, 13, 1522–1532. [Google Scholar]
  216. Amini, A.; Masoumi-Moghaddam, S.; Ehteda, A.; Liauw, W.; Morris, D.L. Potentiation of chemotherapeutics by bromelain and N-acetylcysteine: Sequential and combination therapy of gastrointestinal cancer cells. Am. J. Cancer Res. 2016, 6, 350–369. [Google Scholar]
  217. Mekkawy, A.H.; Breakeit, M.; Pillai, K.; Badar, S.; Akhter, J.; Valle, S.J.; Morris, D.L. Intraperitoneal BromAc((R)) Does Not Interfere with the Healing of Colon Anastomosis. Cancers 2023, 15, 3321. [Google Scholar] [CrossRef]
  218. Dhandayuthapani, S.; Perez, H.D.; Paroulek, A.; Chinnakkannu, P.; Kandalam, U.; Jaffe, M.; Rathinavelu, A. Bromelain-induced apoptosis in GI-101A breast cancer cells. J. Med. Food 2012, 15, 344–349. [Google Scholar] [CrossRef]
  219. Pakbin, B.; Dibazar, S.P.; Allahyari, S.; Shariatifar, H.; Bruck, W.M.; Farasat, A. ACE2-Inhibitory Effects of Bromelain and Ficin in Colon Cancer Cells. Medicina 2023, 59, 301. [Google Scholar] [CrossRef] [PubMed]
  220. Rodriguez-Ortiz, L.; Vazquez-Borrego, M.C.; Bura, F.I.; Granados-Rodriguez, M.; Valenzuela-Molina, F.; Rufian-Andujar, B.; Martinez-Lopez, A.; Ortega-Salas, R.; Espejo-Herrero, J.J.; Romero-Ruiz, A.; et al. Intra-tumoural bromelain and N-acetylcysteine for recurrent and unresectable pseudomyxoma peritonei: Phase I/II trial. Br. J. Surg. 2024, 111, znae045. [Google Scholar] [CrossRef]
Figure 1. Timeline highlighting the key events in the discovery and development of bromelain.
Figure 1. Timeline highlighting the key events in the discovery and development of bromelain.
Nutrients 16 02060 g001
Table 1. Summary of the main features of fruit bromelain and stem bromelain.
Table 1. Summary of the main features of fruit bromelain and stem bromelain.
Fruit Bromelain
(EC 3.4.22.33)
Stem Bromelain
(EC 3.4.22.32)
Source and extraction It is primarily extracted from the fruit (particularly the core) of the pineapple plant. It is obtained by crushing or juicing the fruit and then separating the bromelain enzyme from other components. It is extracted from the stems of the pineapple plant. The stems contain a higher concentration of bromelain compared to the fruit, and the extraction process involves grinding or macerating the stems to release the enzyme.
CompositionTypically contains a mix of proteolytic enzymes, including various cysteine proteases, such as stem bromelain, ananain, and comosain. It may also contain other enzymes and bioactive compound.It consists mainly of cysteine proteases, with the predominant enzyme being bromelain. It may also contain trace amounts of other proteolytic enzymes.
Enzymatic actionHydrolysis of proteins with broad specificity for peptide bonds. Bz-Phe-Val-Arg-/-NHMec is a good synthetic substrate, but there is no action on Z-Arg-Arg-NHMec.Broad specificity for cleavage of proteins but strong preference for Z-Arg-Arg-/-NHMec amongst small molecule substrates.
Table 2. Biological activities and main applications of fruit bromelain and stem bromelain.
Table 2. Biological activities and main applications of fruit bromelain and stem bromelain.
Fruit Bromelain
(EC 3.4.22.33)
Stem Bromelain
(EC 3.4.22.32)
Biological activities
  • Proteolytic activity
  • Anti-inflammatory effects
  • Immunomodulatory effects
  • Antioxidant effects
  • Enzymatic properties
  • Anti-inflammatory effects
  • Immunomodulatory effects
  • Antioxidant effects
  • Promotion of wound healing
  • It may also aid in digestion
Main applications
  • Food processing (e.g., meat tenderizer)
  • Alternative medicine practices
  • Skincare products
  • Food processing (e.g., meat tenderizer)
  • Pharmaceuticals
  • Cosmetics
  • It is often preferred for its higher enzymatic activity and purity.
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

Kansakar, U.; Trimarco, V.; Manzi, M.V.; Cervi, E.; Mone, P.; Santulli, G. Exploring the Therapeutic Potential of Bromelain: Applications, Benefits, and Mechanisms. Nutrients 2024, 16, 2060. https://doi.org/10.3390/nu16132060

AMA Style

Kansakar U, Trimarco V, Manzi MV, Cervi E, Mone P, Santulli G. Exploring the Therapeutic Potential of Bromelain: Applications, Benefits, and Mechanisms. Nutrients. 2024; 16(13):2060. https://doi.org/10.3390/nu16132060

Chicago/Turabian Style

Kansakar, Urna, Valentina Trimarco, Maria V. Manzi, Edoardo Cervi, Pasquale Mone, and Gaetano Santulli. 2024. "Exploring the Therapeutic Potential of Bromelain: Applications, Benefits, and Mechanisms" Nutrients 16, no. 13: 2060. https://doi.org/10.3390/nu16132060

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