Open access peer-reviewed chapter

Boswellia Carries Hope for Patients with Inflammatory Bowel Disease (IBD)

Written By

Sally Elnawasany

Submitted: 12 June 2023 Reviewed: 19 June 2023 Published: 08 July 2023

DOI: 10.5772/intechopen.112244

From the Edited Volume

Medicinal Plants - Chemical, Biochemical, and Pharmacological Approaches

Edited by Mozaniel Santana de Oliveira, Eloisa Helena de Aguiar Andrade, Ravendra Kumar and Suraj N. Mali

Chapter metrics overview

152 Chapter Downloads

View Full Metrics

Abstract

Boswellia serrata is an ancient and valuable herb that was widely used throughout the centuries. Boswellia trees grow in India, Northern Africa, and the Middle East from which Frankincense or olibanum resin is taken. The beneficial effects of Boswellia and its active ingredients (Boswellic acids) were thoroughly investigated in many diseases. Where the non-redox and 5-lipoxygenase inhibitory actions were reported. Inflammatory bowel disease (IBD) mainly ulcerative colitis (UC) and Crohn’s disease (CD) are chronic inflammatory disorders of the gastrointestinal system. Although the cause is still unclear, the immune system is claimed to have the upper hand in the pathogenesis of IBD. Several studies have demonstrated the ameliorating effect of Boswellic acids on the severity of IBD and the potential role of Boswellia in the induction or maintenance of remission. The aim of this chapter is to explore the the possible effect of Boswellia in IBD management as a complementary and alternative strategy.

Keywords

  • Boswellia
  • complementary
  • alternative medicine
  • inflammatory bowel disease
  • ulcerative colitis
  • Crohn’s disease

1. Introduction

Inflammatory bowel disease (IBD) is a chronic inflammatory state classified mainly into ulcerative colitis (UC) and Crohn’s disease (CD). IBD is associated with abdominal pain, diarrhea and rectal bleeding [1]. The choice of treatment for IBD varies according to the type and severity. A variety of medications such as aminosalicylates, corticosteroids, immunosuppressive and biologic drugs are involved [2]. Despite their crucial therapeutic role, some medications carry the risk of infection and cancer [3]. In addition, these drugs are taken throughout life, which leads to patient incompliance and hence treatment failure [4]. These obstacles in treatment insist on the search for other treatments, safe and effective as alternative and complementary modalities.

Advertisement

2. Pathogenesis of IBD

The exact mechanism of IBD is not completely clear [5]. The interaction between genetic factors, changes in intestinal flora homeostasis, environmental variabilities, and intestinal hyperimmune response results in chronic intestinal inflammation [6]. Multiple inflammatory mediators are involved in IBD pathogenesis including leukotrienes, cytokines, chemokines, and prostaglandins. Alteration in reactive oxygen and nitrogen species production adds to the pathogenesis [7]. Based on this, attenuation of hyper-stimulated immune response is the target of IBD therapy. Where the treatment passes in 2 ways, the first is to induce remission and the second is to maintain the remission and to ameliorate intestinal chronic inflammation [8]. In normal conditions, there is a balance among many cells in intestinal lamina propria such as macrophages mast cells, neutrophils, dendritic cells (DCs), eosinophils, natural killer (NK), NKT cells, T and B cells. This provides intestinal protection and tolerance. As a response to bacterial infection, the innate immunity cells (macrophages and dendritic cells) upregulate chemokines and cytokines and act as Antigen-presenting cells (APCs) where they feature the microorganism’s molecular patterns via toll-like receptors (TLR) [9, 10]. Dendritic cells generate native T cell differentiation in mesenteric lymph nodes [11]. With subsequent generation of T helper (Th) subtypes according to the cytokines produced by APCs [12, 13, 14, 15]. Besides proinflammatory cytokines induction, neutrophils stimulate oxidative reactions in the intestinal mucosa [16]. In IBD activity, there is over-expression of many chemokines including macrophage inflammatory proteins (MIP), and Interleukin-8 (IL-8). Under the control of these chemokines, leukocytes are recruited to the inflamed intestine with subsequent oxidative stress [17]. Recruitment of granulocytes and lymphocytes is mediated by adhesion molecules in IBD, such as the intercellular adhesion molecule-1 (ICAM-1), the vascular cell adhesion molecule- (VCAM-) 1, P and E-selectins [18]. In addition, T-cell differentiation and regulation are mediated by cytokines. Th1 differentiation is controlled by IL-12, IL-18, and IL-23 while TNF-α, IL-1β, and IL-6, magnify the immune response by releasing more chemokines and attracting more inflammatory cells [19]. On the other hand, the under-production of IL-10 and Transforming growth factor beta (TGF-β), which are inflammatory attenuating cytokines, contributes to IBD pathogenesis [20, 21].

Advertisement

3. Complementary and alternative (CAM) treatment of IBD

There is a growing worldwide interest in complementary and alternative remedies in IBD treatment [22, 23]. The use of CAM is common for IBD children and adult patients [24, 25]. CAM is found to be commonly used among young, females, at a high educational level or with medication adverse effects [26, 27]. Patients who received massive corticosteroid therapy [28] or suffering from extraintestinal manifestations [24] are more inclined to CAM remedies, as well. Many alternative modalities have been tried in IBD patients including herbs, probiotics, acupuncture and hypnotherapy [8]. Phytochemicals are popularly utilized because of their safety and effectiveness on IBD patients [4, 29]. The variable active herbal ingredients which act on multiple inflammatory pathways and mediators support this preference [25]. Aloe vera, Artemisia absinthium, Boswellia serrata and Curcuma longa were widely studied for their effect on IBD [30].

Advertisement

4. Boswellia serrata

4.1 Structure of Boswellia serrata

Boswellia serrata oleo-gum resin, Indian frankincense was widely used for centuries in traditional medicine. Antioxidant and anti-inflammatory actions have been extensively investigated in several studies on different diseases like colitis, bronchial asthma, arthritis and malignancies [31, 32, 33, 34, 35]. Boswellia serrata resin is composed of monoterpenes, diterpenes, triterpenes, pentacyclic triterpenic acids (boswellic acids) and tetracyclic triterpenic acids [36, 37, 38, 39]. Boswellic acids (BA) compose up 30% of the resin of Boswellia serrata. They are organic acids, formed of a pentacyclic triterpene, a carboxyl group and at least one other functional group [40]. Among boswellic acids, 11-keto-β-boswellic acid (KBA) and acetyl-11-keto-β-boswellic acid (AKBA) are the most active [41].

4.2 Pharmacological activities of Boswellia serrata

4.2.1 Anti-inflammatory action

Boswellia has variable pharmacological activities, anti-inflammatory properties were widely investigated in many studies. Acetyl-boswellic acids block leukotriene production through the downregulation of enzyme 5-lipoxygenase (5-LOX) mediated by a non-redox reaction [42, 43]. In a double-blind placebo control clinical study alcohol extract was given in a 300 mg thrice daily dose for 6 weeks. 70% of asthmatic patients gained clinical improvement [44]. Similarly, gradual control of asthma, regarding the frequency of attacks, pulmonary function tests improvement, and lowering levels of leukotrienes were obtained by another study [45]. In addition, Boswellia ameliorated the inflammation in arthritis [46, 47, 48]. Anti-anaphylactic and mast cell stabilizing effects were also reported where Boswellia suppressed mast cell degranulation [49]. Moreover, boswellic acids were found to possess anti-complement activity [50]. Roy et al. studied the genetic basis of anti-inflammatory effect of BA. Tumor necrosis factor alpha (TNFa) is one of the most crucial mediators of inflammation. TNF alpha induces inflammation by multiple mechanisms, one of them is by upregulation of the expression of adhesion molecules such as microvascular cellular adhesive molecul-1, VCAM-1 in a system of TNF alpha-induced gene expression in human microvascular endothelial cells (HMEC). Of 522 genes that were induced by TNFa 113 genes were highly sensitive to BE treatment both in vivo and in vitro. The function of these genes is linked to inflammation, and cell adhesion [51].

4.2.2 Anti-microbial action

Boswellia oils showed anti-microbial activity against five organisms. Minimum inhibition concentration ranged from 4 to 16 mg/ml against Staphylococcus aureus, 1.5–8.3 mg/ml against Bacillus cereus, 4.0–12.0 mg/ml against Escherichia coli, 2.0–12.8 mg/ml against Proteus vulgaris and 5.3–12.0 mg/ml against Candida albicans [52]. This effect may help in controlling intestinal infection in IBD management.

4.2.3 Anti-tumor action

Boswellic acids induced apoptosis through the upregulation of caspase-8 in colon cancer HT-29 cells [53]. In another study, Boswellia extract altered DNA methylation in colon cancer cells [54]. 4-Amino analogues prepared from β-boswellic acid and 11-keto-β-boswellic acid showed an apoptotic activity mediated by DNA fragmentation [55]. There are multiple pathways by which Boswellia exerts its anti-tumor action such as suppression of topoisomerases I and II [56]. It was also revealed that acetyl keto beta boswellic acid (AKBA) inhibits phosphorylation of ERK pathways and consequently, impairs signal transduction and tumorigenesis [57]. Oxidative stress and nitric oxide production were mediated by Boswellia with subsequent apoptosis in human leukemia HL-60 cells [58]. This anti-tumor potential of Boswellia protects against colorectal cancer which is a common sequala of IBD [59].

4.2.4 Hepato-protective action

In the models of liver injury, hexane extract of oleo-gum-resin of Boswellia in lower doses (87.5 mg/kg p.o.) reduced marker enzymes and prevented the increase in liver weight with histological evidence of hepatoprotection while a mild effect was obtained by higher dose (175 mg/kg p.o.) [60].

4.2.5 Anti-lipidemic action

The aqueous extract of Boswellia carterii with other herbs improved the lipid profile of alloxan-induced diabetic rats [61]. Boswellia showed therapeutic potential for metabolic syndrome. Where Boswellia succeeded in lowering the lipid profile by decreasing the level of TNF-α, IL-1β and increasing the adiponectin level. This action is based on its antioxidant activity [62].

4.2.6 Hypoglycemic action

Herbal formulation containing B. serrata oleo-gum-resin induced a significant anti-diabetic activity on non-insulin-dependent diabetes mellitus [63]. Furthermore, a significant reduction in blood glucose levels and HbA1c was observed when Boswellia serrata was introduced for 17 days to diabetic rats in another study [64].

Based on the mentioned pharmacological actions, Boswellia can help in IBD in many ways, 5-lipoxygenase (5-LO) suppression, downregulation of Tumor necrosis factor alpha (TNF-α) and interleukins, P-selectin-mediated recruitment of inflammatory cells, decreasing reactive oxygen species (ROS), and by modulation intestinal motility [65, 66, 67, 68].

4.3 Preclinical studies of Boswellia in IBD

In vitro study demonstrated the role of Boswellia in the suppression of leukotriene synthesis through interfering with 5-lipoxygenase pathway [69]. The anti-inflammatory effects of boswellic acids extended to inhibit the nuclear transcription factor kappa B (NF-κB) activation, this factor expresses and potentiates the proinflammatory cytokines including TNFα, IL-1β, and IL-6 [70, 71]. This effect was confirmed in an in vitro experimental model of intestinal inflammation where pretreatment of Caco-2 cells monolayers by Boswellia serrata oleo-gum extract (BSE) and AKBA abolished nuclear factor kappa B (NF-κB) activation, protected against cellular changes and inhibited reactive oxygen species (ROS) [72]. Furthermore, the anti-inflammatory and antioxidant properties of Boswellia serrata were explored in acetic acid (AA) induced UC rat model. Daily administration of 34.2 mg/kg of Boswellia serrata extract pre- and post-induction of colitis significantly improved tissue lesions, decreased lipid peroxidation and nitric oxide [73]. A semisynthetic form of acetyl-11-keto-β-boswellic acid ameliorated the disease activity and histology in dextran sodium sulfate (DSS) induced murine colitis. This action was mediated by attenuation of adherent leukocytes and platelets into inflamed tissue by blocking P-selectin stimulation [66]. In an in vitro model of intestinal inflammation, the anti-inflammatory action of Boswellia serrata and C. longa were tested. Boswellia serrata at 1 μg/mL protected the intestinal epithelium with a 25% reduction of ROS generation [74]. The immunosuppressive potential of Boswellia carteri gum resin extract was illuminated in an in vitro study, where the extract attenuated human primary T lymphocyte proliferation in a concentration-dependent manner via nuclear factor of activated T-cells (NFAT) dependent mechanism [75]. Metabolism of sphingomyelin induces lipid signals that impact cell proliferation, and inflammation. 3-acetyl-11-keto-β-boswellic acids (AKBA) attenuated the expression of sphingomyelinase in intestinal cells. This provides other anti-inflammatory mechanism of Boswellia [76]. A new herbal formulation that includes Punica granatum L, Boswellia serrata, and Curcuma longa L extracts, inhibited TNF-alfa-induced release of IL-8, IL-6 and Monocyte chemoattractant protein-1 (MCP-1) in Caco-2 cells [77]. Although many studies support the anti-inflammatory action of Boswellia, on the contrary, Kiela et al. claimed that Boswellia showed no effect on improving colitis in dextran sulfate sodium (DSS)- or trinitrobenzene sulfonic acid- (TNBS-) induced experimental models [78]. Anti-fibrotic effect of Boswellia was declared when oral Boswellia in combination with significantly improved the inflammation of trinitrobenzene sulphonic acid (TNBS)-induced chronic colitis. There was an improvement in the histological features of colonic fibrosis. Together with a significant reduction in the expression of alpha-smooth muscle actin (α-SMA), collagen I–III, connective tissue growth factor (CTGF) and transforming growth factor-beta1 (TGF-β1) [79].

4.4 Clinical studies of Boswellia in IBD

Boswellia serrata in a dose of 350 mg thrice daily for 6 weeks achieved remission in 82% of treated UC patients compared to 75% of patients who were treated with sulfasalazine (1 g thrice daily). Inflammatory parameters were better in Boswellia treated group as well [80]. Another trial was conducted on 30 patients with chronic colitis for 6 weeks. Of 20 patients who received daily 900 mg Boswellia serrata, improvement of inflammatory parameters and remission were noticed in (18 and 14 patients, respectively) while after treatment of 10 patients with 3 gm sulfasalazine daily 6 participants showed improvement in inflammatory parameters and remission was achieved in 4 patients [81]. Boswellia tolerability and ability to maintain remission were demonstrated in a 52-week multicenter double-blind, placebo-controlled, randomized Germain study. 82 CD patients were randomly divided, 42 patients received daily Boswellia in 400 mg capsules and 40 patients received a placebo. There was no difference in both groups in parameters of inflammation or disease activity or maintaining remission and Boswellia was well tolerated [82]. Another 4-week trial was conducted on UC patients who were in remission. Boswellia serrata extract (BSE) was introduced orally, in a novel delivery form to 22 patients compared to 21 patients with no treatment. Improvement in clinical parameters, a decrease in medication needs and a lowering in fecal calprotectin levels were observed in Boswellia treated group [83]. The effect of Boswellia was tried in patients with collagenous colitis as well, where 400 mg oral BSE was given thrice daily for six weeks compared to a placebo. The remission rate was higher in BSE-treated patients but without any changes in histology or quality of life [34]. We conducted a 6-week clinical trial on 60 patients with active ulcerative colitis to investigate the effect of Boswellia extract on disease activity. 20 patients received Mesalamine 3, 20 patients received Boswellia extract in the form of oral tablets in a dose of 2 gm/day, and 20 patients were given Boswelliaextract plus Mesalamine in the mentioned doses. Clinical and laboratory improvement was noticed in the three groups without a significant difference. There were no recorded side effects of Boswellia during the 6 weeks of the study [84].

Advertisement

5. Conclusion

Based on preclinical and clinical studies, the anti-inflammatory, immune modulation, and anti-cancer activities of Boswellia as well as its safety and tolerability recommend its therapeutic use in IBD patients.

Advertisement

Acknowledgments

I thank God Almighty for his countless blessings, including Boswellia, and I thank my patients for their patience and trust. I extend my gratitude to Professor Dr. Farid Badria for his continuous support.

Advertisement

Conflict of interest

I confirm that there are no conflicts of interest.

References

  1. 1. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: Clinical aspects and established and evolving therapies. The Lancet. 2007;369(9573):1641-1657
  2. 2. Seyedian SS, Nokhostin F, Malamir MD. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. Journal of Medicine and Life. 2019;12(2):113-122
  3. 3. Toruner M, Loftus EV Jr, Harmsen WS, Zinsmeister AR, Orenstein R, Sandborn WJ, et al. Risk factors for opportunistic infections in patients with inflammatory bowel disease. Gastroenterology. 2008;134(4):929-936
  4. 4. Ng S, Lam Y, Tsoi K, Chan F, Sung J, Wu J. Systematic review: The efficacy of herbal therapy in inflammatory bowel disease. Alimentary Pharmacology & Therapeutics. 2013;38(8):854-863
  5. 5. Haag L-M, Siegmund B. Exploring & exploiting our ‘other self’–does the microbiota hold the key to the future therapy in Crohn's? Best Practice & Research Clinical Gastroenterology. 2014;28(3):399-409
  6. 6. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140(6):1756-67. e1
  7. 7. Xavier RJ, Podolsky DK. Unravelling the pathogenesis of inflammatory bowel disease. Nature. 2007;448(7152):427-434
  8. 8. Bernstein CN. Treatment of IBD: Where we are and where we are going. Official Journal of the American College of Gastroenterology| ACG. 2015;110(1):114-126
  9. 9. Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clinical Microbiology Reviews. 2009;22(2):240-273
  10. 10. Smith P, Smythies L, Shen R, Greenwell-Wild T, Gliozzi M, Wahl S. Intestinal macrophages and response to microbial encroachment. Mucosal Immunology. 2011;4(1):31-42
  11. 11. Cerovic V, Houston S, Scott C, Aumeunier A, Yrlid U, Mowat A, et al. Intestinal CD103− dendritic cells migrate in lymph and prime effector T cells. Mucosal Immunology. 2013;6(1):104-113
  12. 12. Usui T, Preiss JC, Kanno Y, Yao ZJ, Bream JH, O'Shea JJ, et al. T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription. The Journal of Experimental Medicine. 2006;203(3):755-766
  13. 13. Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. Journal of Biological Chemistry. 2007;282(13):9358-9363
  14. 14. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1β and 6 but not transforming growth factor-β are essential for the differentiation of interleukin 17–producing human T helper cells. Nature Immunology. 2007;8(9):942-949
  15. 15. Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, et al. Development, cytokine profile and function of human interleukin 17–producing helper T cells. Nature Immunology. 2007;8(9):950-957
  16. 16. Seguí J, Gironella M, Sans M, Granell S, Gil F, Gimeno M, et al. Superoxide dismutase ameliorates TNBS-induced colitis by reducing oxidative stress, adhesion molecule expression, and leukocyte recruitment into the inflamed intestine. Journal of Leukocyte Biology. 2004;76(3):537-544
  17. 17. Laing KJ, Secombes CJ. Chemokines. Developmental & Comparative Immunology. 2004;28(5):443-460
  18. 18. Vainer B. Intercellular adhesion molecule-1 (ICAM-1) in ulcerative colitis: Presence, visualization, and significance. APMIS. 2010;118:1-46
  19. 19. Sanchez-Muñoz F, Dominguez-Lopez A, Yamamoto-Furusho JK. Role of cytokines in inflammatory bowel disease. World Journal of Gastroenterology: WJG. 2008;14(27):4280
  20. 20. Izcue A, Coombes JL, Powrie F. Regulatory lymphocytes and intestinal inflammation. Annual Review of Immunology. 2009;27:313-338
  21. 21. Li MO, Flavell RA. Contextual regulation of inflammation: A duet by transforming growth factor-β and interleukin-10. Immunity. 2008;28(4):468-476
  22. 22. Joos S, Rosemann T, Szecsenyi J, Hahn EG, Willich SN, Brinkhaus B. Use of complementary and alternative medicine in Germany–a survey of patients with inflammatory bowel disease. BMC Complementary and Alternative Medicine. 2006;6:1-7
  23. 23. Hilsden RJ, Verhoef MJ, Best A, Pocobelli G. Complementary and alternative medicine use by Canadian patients with inflammatory bowel disease: Results from a national survey. The American Journal of Gastroenterology. 2003;98(7):1563-1568
  24. 24. Fernández A, Barreiro-de Acosta M, Vallejo N, Iglesias M, Carmona A, González-Portela C, et al. Complementary and alternative medicine in inflammatory bowel disease patients: Frequency and risk factors. Digestive and Liver Disease. 2012;44(11):904-908
  25. 25. Hilsden RJ, Verhoef MJ, Rasmussen H, Porcino A, DeBruyn JC. Use of complementary and alternative medicine by patients with inflammatory bowel disease. Inflammatory Bowel Diseases. 2011;17(2):655-662
  26. 26. Opheim R, Hoivik ML, Solberg IC, Moum B, Group IS. Complementary and alternative medicine in patients with inflammatory bowel disease: The results of a population-based inception cohort study (IBSEN). Journal of Crohn's and Colitis. 2012;6(3):345-353
  27. 27. Opheim R, Bernklev T, Fagermoen MS, Cvancarova M, Moum B. Use of complementary and alternative medicine in patients with inflammatory bowel disease: Results of a cross-sectional study in Norway. Scandinavian Journal of Gastroenterology. 2012;47(12):1436-1447
  28. 28. Langhorst J, Anthonisen IB, Steder-Neukamm U, Lüdtke R, Spahn G, Michalsen A, et al. Amount of systemic steroid medication is a strong predictor for the use of complementary and alternative medicine in patients with inflammatory bowel disease. Results from a German national survey. Inflammatory Bowel Diseases. 2005;11(3):287-295
  29. 29. Ke F, Yadav PK, Ju LZ. Herbal medicine in the treatment of ulcerative colitis. Saudi Journal of Gastroenterology: Official Journal of the Saudi Gastroenterology Association. 2012;18(1):3
  30. 30. Algieri F, Rodriguez-Nogales A, Rodriguez-Cabezas ME, Risco S, Ocete MA, Galvez J. Botanical drugs as an emerging strategy in inflammatory bowel disease: A review. Mediators of Inflammation. 2015;2015:179616
  31. 31. Moussaieff A, Mechoulam R. Boswellia resin: From religious ceremonies to medical uses; a review of in-vitro, in-vivo and clinical trials. Journal of Pharmacy and Pharmacology. 2009;61(10):1281-1293
  32. 32. Siddiqui MZ. Boswellia serrata, a potential antiinflammatory agent: An overview. Indian Journal of Pharmaceutical Sciences. 2011;73(3):255
  33. 33. Ernst E. Frankincense: systematic review. British Medical Journal. 2008;337:a2813
  34. 34. Madisch A, Miehlke S, Eichele O, Mrwa J, Bethke B, Kuhlisch E, et al. Boswellia serrata extract for the treatment of collagenous colitis. A double-blind, randomized, placebo-controlled, multicenter trial. International journal of colorectal disease. 2007;22:1445-1451
  35. 35. Huang MT, Badmaev V, Ding Y, Liu Y, Xie JG, Ho CT. Anti-tumor and anti-carcinogenic activities of triterpenoid, β-boswellic acid. BioFactors. 2000;13(1-4):225-230
  36. 36. El Khadem H, El-Shafei Z, El Sekeily M, Rahman MA. Derivatives of boswellic acids. Planta Medica. 1972;22(06):157-159
  37. 37. Hamm S, Bleton J, Connan J, Tchapla A. A chemical investigation by headspace SPME and GC–MS of volatile and semi-volatile terpenes in various olibanum samples. Phytochemistry. 2005;66(12):1499-1514
  38. 38. Safayhi H, Mack T, Sabieraj J, Anazodo MI, Subramanian LR, Ammon H. Boswellic acids: novel, specific, nonredox inhibitors of 5-lipoxygenase. Journal of Pharmacology and Experimental Therapeutics. 1992;261(3):1143-1146
  39. 39. Mahajan B, Taneja S, Sethi V, Dhar K. Two triterpenoids from Boswellia serrata gum resin. Phytochemistry. 1995;39(2):453-455
  40. 40. Ammon H. Boswellic acids (components of frankincense) as the active principle in treatment of chronic inflammatory diseases. Wiener Medizinische Wochenschrift (1946). 2002;152(15-16):373-378
  41. 41. Abdel-Tawab M, Werz O, Schubert-Zsilavecz M. Boswellia serrata: An overall assessment of in vitro, preclinical, pharmacokinetic and clinical data. Clinical Pharmacokinetics. 2011;50:349-369
  42. 42. Krieglstein CF, Anthoni C, Rijcken EJ, Laukötter M, Spiegel H-U, Boden SE, et al. Acetyl-11-keto-β-boswellic acid, a constituent of a herbal medicine from Boswellia serrata resin, attenuates experimental ileitis. International Journal of Colorectal Disease. 2001;16:88-95
  43. 43. Ammon H, Safayhi H, Mack T, Sabieraj J. Mechanism of antiinflammatory actions of curcumine and boswellic acids. Journal of Ethnopharmacology. 1993;38(2-3):105-112
  44. 44. Gupta I, Gupta V, Parihar A, Gupta S, Lüdtke R, Safayhi H, et al. Effects of Boswellia serrata gum resin in patients with bronchial asthma: Results of a double-blind, placebo-controlled, 6-week clinical study. European Journal of Medical Research. 1998;3(11):511-514
  45. 45. Badria FA, Mohammed EA, El-Badrawy MK, El-Desouky M. Natural leukotriene inhibitor from Boswellia: A potential new alternative for treating bronchial asthma. Alternative & Complementary Therapies. 2004;10(5):257-265
  46. 46. Badria FA, El-Farahaty T, Shabana AA, Hawas SA, El-Batoty MF. Boswellia–curcumin preparation for treating knee osteoarthritis: A clinical evaluation. Alternative & Complementary Therapies. 2002;8(6):341-348
  47. 47. Bannuru RR, Osani MC, Al-Eid F, Wang C. Efficacy of curcumin and Boswellia for knee osteoarthritis: Systematic review and meta-analysis. Seminars in Arthritis and Rheumatism. 2018;48(3):416-429
  48. 48. Singh S, Khajuria A, Taneja SC, Khajuria RK, Singh J, Qazi GN. Boswellic acids and glucosamine show synergistic effect in preclinical anti-inflammatory study in rats. Bioorganic & Medicinal Chemistry Letters. 2007;17(13):3706-3711
  49. 49. Pungle P, Banavalikar M, Suthar A, Biyani M, Mengi S. Immunomodulatory activity of boswellic acids of Boswellia serrata Roxb. Indian journal of experimental biology. 2003;41(12):1460-1462
  50. 50. Kapil A, Moza N. Anticomplementary activity of boswellic acids--an inhibitor of C3-convertase of the classical complement pathway. International Journal of Immunopharmacology. 1992;14(7):1139-1143
  51. 51. Roy S, Khanna S, Shah H, Rink C, Phillips C, Preuss H, et al. Human genome screen to identify the genetic basis of the anti-inflammatory effects of Boswellia in microvascular endothelial cells. DNA and Cell Biology. 2005;24(4):244-255
  52. 52. Van Vuuren SF, Kamatou GP, Viljoen AM. Volatile composition and antimicrobial activity of twenty commercial frankincense essential oil samples. South African Journal of Botany. 2010;76(4):686-691
  53. 53. Liu J-J, Nilsson Å, Oredsson S, Badmaev V, Zhao W-Z, Duan R-D. Boswellic acids trigger apoptosis via a pathway dependent on caspase-8 activation but independent on Fas/Fas ligand interaction in colon cancer HT-29 cells. Carcinogenesis. 2002;23(12):2087-2093
  54. 54. Shen Y, Link A, Takahashi M, Balaguer F, Hur K, Boland CR, et al. Boswellia extracts induce DNA methylation changes in colon cancer cells. Gastroenterology. 2011;5(140):S-400
  55. 55. Shah BA, Kumar A, Gupta P, Sharma M, Sethi VK, Saxena AK, et al. Cytotoxic and apoptotic activities of novel amino analogues of boswellic acids. Bioorganic & Medicinal Chemistry Letters. 2007;17(23):6411-6416
  56. 56. Syrovets T, Büchele B, Gedig E, Slupsky JR, Simmet T. Acetyl-boswellic acids are novel catalytic inhibitors of human topoisomerases I and IIα. Molecular Pharmacology. 2000;58(1):71-81
  57. 57. Park YS, Lee JH, Bondar J, Harwalkar JA, Safayhi H, Golubic M. Cytotoxic action of acetyl-11-keto-β-boswellic acid (AKBA) on meningioma cells. Planta Medica. 2002;68(05):397-401
  58. 58. Bhushan S, Kumar A, Malik F, Andotra SS, Sethi VK, Kaur IP, et al. A triterpenediol from Boswellia serrata induces apoptosis through both the intrinsic and extrinsic apoptotic pathways in human leukemia HL-60 cells. Apoptosis. 2007;12:1911-1926
  59. 59. Lucafò M, Curci D, Franzin M, Decorti G, Stocco G. Inflammatory bowel disease and risk of colorectal cancer: An overview from pathophysiology to pharmacological prevention. Frontiers in Pharmacology. 2021;12:772101
  60. 60. Jyothi Y, Kamath JV, Asad M. Effect of hexane extract of Boswellia serrata oleo-gum resin on chemically induced liver damage. Pakistan Journal of Pharmaceutical Sciences. 2006;19(2):125-129
  61. 61. Helal EG, Shahat M. Hypolipidimic effect of some medicinal plants on diabetic rats. The Egyptian Journal of Hospital Medicine. 2006;23(1):200-211
  62. 62. Mahdian D, Abbaszadeh-Goudarzi K, Raoofi A, Dadashizadeh G, Abroudi M, Zarepour E, et al. Effect of Boswellia species on the metabolic syndrome: A review. Iranian Journal of Basic Medical Sciences. 2020;23(11):1374-1381
  63. 63. Al-Awadi F, Fatania H, Shamte U. The effect of a plants mixture extract on liver gluconeogenesis in streptozotocin induced diabetic rats. Diabetes Research (Edinburgh, Scotland). 1991;18(4):163-168
  64. 64. Azemi ME, Namjoyan F, Khodayar MJ, Ahmadpour F, Padok AD, Panahi M. The antioxidant capacity and anti-diabetic effect of Boswellia serrata Triana and planch aqueous extract in fertile female diabetic rats and the possible effects on reproduction and histological changes in the liver and kidneys. Jundishapur Journal of Natural Pharmaceutical Products. 2012;7(4):168-175
  65. 65. Poeckel D, Werz O. Boswellic acids: Biological actions and molecular targets. Current Medicinal Chemistry. 2006;13(28):3359-3369
  66. 66. Anthoni C, Laukoetter MG, Rijcken E, Vowinkel T, Mennigen R, Muller S, et al. Mechanisms underlying the anti-inflammatory actions of boswellic acid derivatives in experimental colitis. American journal of physiology-gastrointestinal and liver. Physiology. 2006;290(6):G1131-G11G7
  67. 67. Ammon H. Modulation of the immune system by Boswellia serrata extracts and boswellic acids. Phytomedicine. 2010;17(11):862-867
  68. 68. Borrelli F, Capasso F, Capasso R, Ascione V, Aviello G, Longo R, et al. Effect of Boswellia serrata on intestinal motility in rodents: Inhibition of diarrhoea without constipation. British Journal of Pharmacology. 2006;148(4):553
  69. 69. Stanke-Labesque F, Pofelski J, Moreau-Gaudry A, Bessard G, Bonaz B. Urinary leukotriene E4 excretion: A biomarker of inflammatory bowel disease activity. Inflammatory Bowel Diseases. 2008;14(6):769-774
  70. 70. Takada Y, Ichikawa H, Badmaev V, Aggarwal BB. Acetyl-11-keto-β-boswellic acid potentiates apoptosis, inhibits invasion, and abolishes osteoclastogenesis by suppressing NF-κB and NF-κB-regulated gene expression. The Journal of Immunology. 2006;176(5):3127-3140
  71. 71. Wang H, Syrovets T, Kess D, Büchele B, Hainzl H, Lunov O, et al. Targeting NF-κB with a natural triterpenoid alleviates skin inflammation in a mouse model of psoriasis. The Journal of Immunology. 2009;183(7):4755-4763
  72. 72. Catanzaro D, Rancan S, Orso G, Dall'Acqua S, Brun P, Giron MC, et al. Boswellia serrata preserves intestinal epithelial barrier from oxidative and inflammatory damage. PLoS One. 2015;10(5):e0125375
  73. 73. Hartmann RM, Fillmann HS, Morgan Martins MI, Meurer L, Marroni NP. Boswellia serrata has beneficial anti-inflammatory and antioxidant properties in a model of experimental colitis. Phytotherapy Research. 2014;28(9):1392-1398
  74. 74. Governa P, Marchi M, Cocetta V, De Leo B, Saunders PT, Catanzaro D, et al. Effects of Boswellia Serrata Roxb. And Curcuma longa L. in an in vitro intestinal inflammation model using immune cells and Caco-2. Pharmaceuticals. 2018;11(4):126
  75. 75. Zimmermann-Klemd AM, Reinhardt JK, Nilsu T, Morath A, Falanga CM, Schamel WW, et al. Boswellia carteri extract and 3-O-acetyl-alpha-boswellic acid suppress T cell function. Fitoterapia. 2020;146:104694
  76. 76. Zhang Y, Duan R-D. Boswellic acid inhibits expression of acid sphingomyelinase in intestinal cells. Lipids in Health and Disease. 2009;8:1-8
  77. 77. Petti L, De Santis F, Vetrano S. In vitro anti-inflammatory and protective effects of ibidì® on intestinal epithelial cells. European Journal of Medicinal Plants. 2014;4(9):1022
  78. 78. Kiela PR, Midura AJ, Kuscuoglu N, Jolad SD, Sólyom AM, Besselsen DG, et al. Effects of Boswellia serrata in mouse models of chemically induced colitis. American journal of physiology-gastrointestinal and liver. Physiology. 2005;288(4):G798-G808
  79. 79. Latella G, Sferra R, Vetuschi A, Zanninelli G, D’angelo A, Catitti V, et al. Prevention of colonic fibrosis by Boswellia and Scutellaria extracts in rats with colitis induced by 2, 4, 5-trinitrobenzene sulphonic acid. European Journal of Clinical Investigation. 2008;38(6):410-420
  80. 80. Gupta I, Parihar A, Malhotra P, Singh GB, Lüdtke R, Safayhi H, et al. Effects of Boswellia serrata gum resin in patients with ulcerative colitis. European Journal of Medical Research. 1997;2(1):37-43
  81. 81. Gupta I, Parihar A, Malhotra P, Gupta S, Lüdtke R, Safayhi H, et al. Effects of gum resin of Boswellia serrata in patients with chronic colitis. Planta Medica. 2001;67(5):391-395
  82. 82. Holtmeier W, Zeuzem S, Preiβ J, Kruis W, Böhm S, Maaser C, et al. Randomized, placebo-controlled, double-blind trial of Boswellia serrata in maintaining remission of Crohn's disease: Good safety profile but lack of efficacy. Inflammatory Bowel Diseases. 2011;17(2):573-582
  83. 83. Pellegrini L, Milano E, Franceschi F, Belcaro G, Gizzi G, Feragalli B, et al. Managing ulcerative colitis in remission phase: Usefulness of Casperome®, an innovative lecithin-based delivery system of Boswellia serrata extract. European Review for Medical and Pharmacological Sciences. 2016;20(12):2695-2700
  84. 84. Elnawasany S, Assal F, Awad A, Eldesoky K, Badria F. Study of the effect of Boswellia serrata on patients with ulcerative colitis [dissertation for the doctorate]: Tanta University; 2012. Available from: https://tanta.edu.eg

Written By

Sally Elnawasany

Submitted: 12 June 2023 Reviewed: 19 June 2023 Published: 08 July 2023