DiOne
Tab DiOne
(Diacerein, Glucosamine, Chondroitin and MSM)
Brand: DiOne
Generic: Diacerein, Glucosamine, MSM and Chondroitin
Class: Medical Food Supplement
Route of administration: Oral
Dosage form: Tablet
Dose: 1 tablet twice a day
Contraindications: Pregnancy: Cat. C
Pack Size: 30 tabs.
Price: Rs. 960/-
Supplement facts:
Supplement Amount per serving
Diacerein 50mg
Glucosamine 600mg
Chondroitin 400mg
MSM 168mg
1. Diacerein
The use of diacerein for the treatment of osteoarthritis is still under sometimes controversial discussion. Its postulated modes of action, all indicating IL-1 antagonism, make the drug an interesting option for the treatment of osteoarthritis not only for symptom relief but also for structure modification. Studies in different animal models [1-8] showed that diacerein consistently moderated cartilage loss in osteoarthritis. In humans, the ECHODIAH trial [9] showed a reduction in the progression of hip osteoarthritis in the diacerein-treated patients compared with the placebo group.
Overall, this meta-analysis provides evidence for statistically significant and clinically relevant efficacy of diacerein on improvement of pain and function in patients with hip and/or knee osteoarthritis. The large number of patients included in this meta-analysis gives the basis for well-founded conclusions to be drawn. Considering the pain relief results, one has to consider that complete withdrawal of analgesic co-medication was not possible during the studies analyzed. In most of the trials, acetaminophen, which is also considered an appropriate treatment modality for moderate osteoarthritis, [10,11] was allowed as an escape medication in both the diacerein and control groups, and the amount taken was recorded by the patient in a diary. However, although mean intake of acetaminophen was similar during the active treatment period, its consumption increased significantly in the NSAID-treated patients, but not in the diacerein group, during the treatment-free follow-up period. After the end of treatment, SYSADOAs are supposed to have a carryover effect [12] In this meta-analysis, we found evidence for this carryover effect with respect to pain and consumption of escape medication in all sub analyses performed, not only in the individual trials but also in the pooled population, and not only compared with placebo [13,14] but also with active treatment modalities comprising diclofenac sodium, [15,16] tenoxicam, [17] piroxicam,[18,19] and naproxen [20]. Given the fact that all selected studies had a randomized, controlled, parallel-group de-sign, these findings indicate a positive impact of diacerein on the clinical status of patients with osteoarthritis. The patient’s global tolerability ratings at the end of active treatment showed no statistically significant differences between other active treatments and diacerein. However, as expected, a significant inferiority of diacerein vs placebo was observed, also indicating reproducibility of the results obtained here.
The risk-benefit ratio associated with long-term use of NSAIDs and analgesics is well documented in the literature, [20,21-23] but clinical studies on the use of NSAIDs for more than 6 weeks in patients with osteoarthritis are rare [20,24]. Nonsteroidal anti-inflammatory drugs, particularly the selective cyclooxygenase-2 inhibitors, are known to exert a higher risk for thromboembolic disorders such as myocardial infarction or stroke [25]. In this context, it is important to consider that most patients affected by osteoarthritis also experience disorders, or at least risk factors, of the cardiovascular system and that according to the European Agency for the Evaluation of Medicinal Products [26] and the Food and Drug Administration, [27] NSAIDs should be administered at the lowest possible dose for the shortest period. In contrast, cardiovascular adverse events in patients treated with diacerein can be considered very rare. In France, over a period of 11 years (from September 1994 to November 2005) and with more than 14 million prescriptions of diacerein, only 9 cases of cardiovascular adverse events with diacerein were spontaneously reported (information collected by the Drug Safety Department, Negma-Lerads, Toussus-Le-Noble, France). In particular, no acute coronary syndrome or myocardial infarction was reported. Thus, with respect to tolerability, diacerein may have some advantages compared with long-term application of NSAIDs.
2. Glucosamine and Chondroitin
Currently, glucosamine (glucosamine sulfate (GS); glucosamine hydrochloride (GH)) and CS are the most commonly used supplements to ease the pain and discomfort of arthritis in humans and animals. Glucosamine and chondroitin are components of the ECM of articular cartilage. Glucosamine (2-amino-2-deoxy-d-glucose) is an amino sugar and a constituent of glycosaminoglycan (GAG), which plays a role in the normal growth and repair of articular cartilage. In fact, glucosamine is considered a building block of cartilage.
Glucosamine is extracted from crab, lobster, and shrimp shells. Following oral administration of either GS or GH, these salts are ionized in the stomach, making glucosamine available for absorption in the small bowel. Ninety percent of GS is absorbed, but due to an extensive first-pass metabolism, bioavailability approaches only 25% [28] Its excretion is mainly through the kidneys, with only a small unmodified amount eliminated through the stool. CS is a sulfated GAG composed of a chain of alternating sugars, N-acetyl-d-galactosamine and d-glucuronic acid. CS is a normal constituent of aggrecan, the major proteoglycan of articular cartilage. It is extracted from animal cartilage, such as trachea and shark cartilage. Due to its larger size as compared to glucosamine, approximately 30% of CS is absorbed, with 12–13% bioavailability [29]. Studies suggest that glucosamine helps relieve pain by enhancing proteoglycan synthesis, which is impaired in osteoarthritic cartilage [30] . [31] investigated the effects of glucosamine on MMP production, MAPK phosphorylation, and activator protein (AP)-1 transcription factor activation in human chondrocytes. Findings revealed that glucosamine reduced the expression of MMPs (MMP-1, MMP-3, andMMP-13) and inhibited c-jun amino terminal kinase, p38 phosphorylation, and, consequently, c-jun binding activity.
In essence, glucosamine inhibits IL-1β-stimulated MMP production in human chondrocytes by affecting MAPK phosphorylation. The glucosamine and CS combination suppresses IL-1-induced gene expression of iNOS, COX-2, mPGEs, and NF-κB in cartilage explants. This leads to reduced production of NO and PGE2, two mediators responsible for the cell death of chondrocytes and inflammatory reactions [32]. CS provides hydration, assists in cushioning impact stress, helps create osmotic pressure within the ECM to maintain the compressive resistance of cartilage, improves function/mobility of the joint, reduces the progression of OA, and reduces joint pain [33] CS has been shown to increase hyaluronan production by human synovial cells, which has a beneficial effect on maintaining viscosity in the SF. CS stimulates chondrocyte metabolism, leading to the synthesis of collagen and proteoglycan, the basic components of new cartilage [34]. CS also provides elasticity and assists in cushioning impact stress. It is suggested that CS may help the body to repair damaged cartilage and help restore joint integrity. It may also protect existing cartilage from premature breakdown. Because CS production by the body decreases with age, its supplementation may be especially helpful for older humans. Furthermore, CS inhibits the enzymes leukocyte elastase and hyaluronidase, which are found in high concentrations in SF of patients with rheumatic diseases. It has also been hypothesized to reduce inflammation, inhibit synthesis of degradative enzymes including MMPs, increase synthesis of ECM constituents, and reduce apoptosis of articular chondrocytes [35]. [36] reviewed details of the biochemical basis of the effect of CS on OA articular tissue. At molecular levels, CS inhibits NF-κB nuclear translocation and phosphorylation of p38 MAPK, ERK1/2, and JNK [37].
When given in combination, these two supplements: stimulate the synoviocyte and chondrocyte metabolism; inhibit the enzymatic degradation and reduce the fibrin thrombi in the periarticular microcirculation; and can regulate the genetic expression and the synthesis of NO and PGE2, thereby exerting anti-inflammatory properties. In addition to anti-inflammatory action, GS and CS exhibit an antioxidant expression that leads to a significant reduction in iNOS expression and activity [38], thereby reducing the otherwise NO-induced cell death of chondrocytes. In several studies, GS and CS have been evaluated as single agents and in combination for the treatment of OA. In fact, it is a common practice for glucosamine and chondroitin to be used together because they offer a greater beneficial effect than when given alone, although they work through different mechanisms of action. In a clinical trial, GS was found to be as effective or slightly more effective than analgesics [39,40,41,] or NSAIDs [42] for decreasing pain. A trial by [43] did not show a difference between GS and placebo. In another clinical trial,GS was found to be ineffective for reducing pain in patients with severe knee OA, but it was more effective when it was used in combination with CS due to a synergistic effect in patients with moderate to severe pain [44] found that the chondroprotective agents (GS and CS) were effective in improving the function of patients with OA, but the radiological modifications in the knee were statistically insignificant after 12 months of monitoring. However, the findings of Narvy and Vangsness (2010) supported a role of these nutraceuticals in reducing radiographic progression of knee OA. Recently, Kozakcioğlu (2012) revealed that GS and CS both have a suppressor effect on the structural deformities of OA.
In a recent study, Erhan et al. (2012) showed that topical glucosamine treatment combined with physical therapy in patients with knee OA had no superiority over placebo, based on radiological findings (Kellgren– Lawrence score) and joint stiffness index (Western Ontario and McMaster Osteoarthritis, WOMAC score). In all clinical trials, GS, GH, and CS were found to be as safe as placebo [45]. The acute oral LD50 of glucosamine is >8 g/kg in rats and 15 g/kg in mice, which can be considered nontoxic. Oral administration of glucosamine at 2,700 mg/kg for 12 months in animals produced no adverse effects. Oral administration of large doses of glucosamine in animals has no documented effects on glucose metabolism. However, Lafontaine-Lacasse et al. (2011) reported that, at beyond recommended dosages, glucosamine may damage pancreatic cells, thereby possibly increasing the risk of developing diabetes. In a few patients, hepatotoxicity (hepatitis and/or cholestasis) developed following exposure to glucosamine [46] These authors established a temporal relationship between onset of liver injury and glucosamine ingestion.
Clinical studies have not identified any significant side effects of CS, which suggests its long-term safety [47]. Recently, the Task Force of the European League against Rheumatism (EULAR) committee also granted CS a level of toxicity of 6 on a 0–100 scale, confirming that CS is one of the safest remedies for OA. Currently, glucosamine and chondroitin are not recommended by Osteoarthritis Research Society International (OARSI) and the American College of Rheumatology (ACR) [48]. In dogs with moderate OA, daily administration of GH (2,000 mg) and CS (1,600 mg) for a period of 120–150 days significantly reduced pain associated with OA [49]. Gupta et al. (2009) also reported significant reduction of pain in horses with moderate OA receiving GH (5.4 g) and CS (1.8 g) daily for a period of 150 days. In both dogs and horses, GH and CS produced no side effects and were well-tolerated. Previously, in a number of in vivo and in vitro studies, GS and CS have been found to be very effective against OA in animal models [50]. These studies suggested that the combination of GS and CS appears to be more effective in preventing or treating OA in animals than either product alone. A Golden Retriever developed polyuria and polydipsia when treated with 1,000 mg of glucosamine/day [51]. The polyuria and polydipsia resolved after the glucosamine dose was lowered to 500 mg/day. In an experimental study, glucosamine was reported to cause hyperglycemia, insulin resistance, and a diabetic-like state in rats [52]. Beriault et al. did not confirm hyperglycemia and insulin resistance with glucosamine, but it promoted endoplasmic reticulum stress, hepatic steatosis, and accelerated atherogenesis in mice.
3. Methylsylfonylmethane (MSM)
Methylsylfonylmethane (MSM), also known as methyl sulfonate or dimethyl sulfone, is a naturally occurring organosulfur compound and a putative methyl donor. MSM is the first oxidized metabolite of dimethyl sulfoxide (DMSO). It occurs naturally in some plants and it is added in small amounts to many foods and beverages as a dietary supplement. Currently, MSM is sold via 52 different products as a single agent in capsule, caplet, lotion, and cream forms, and in more than 30 different products in combination with other dietary supplements. MSM is sold as a dietary supplement and marketed with a variety of claims often in combination with glucosamine and/or chondroitin for helping to treat or prevent OA. However, there is a paucity of scientific evidence to support the use of MSM. It has been suggested that the benefits claimed for MSM far exceed the number of scientific studies. In October 2000, the US Food and Drug Administration (FDA) warned one MSM promoter, Karl Loren, to cease and desist from making therapeutic claims for MSM. Due to its sulfur content, MSM is used by the body to maintain normal connective tissue. MSM may have anti-inflammatory activity, chemopreventive property, prostacyclin (PGI2) synthesis inhibition, antiatherosclerotic action, salutary effect on eicosanoid metabolism, and free radical scavenging activity [53,54]. MSM is an analgesic, anti-inflammatory, and blood vessel dilator. In OA, MSM works by reducing inflammation and blocking the pain response in nerve fibers. However, like any other nutraceutical, MSM does not cure OA. MSM serves the same purpose as the NSAIDs, but MSM has none of the negative outcomes associated with NSAIDs. In a randomized, double-blind, placebo-controlled trial,[54] reported that compared to placebo, 3 g of MSM twice per day (6 g/day total) for 12 weeks produced significant decreases in WOMAC pain and physical function impairment (P < 0.041 and P < 0.045, respectively). No notable changes were found in WOMAC stiffness and aggregated total symptoms scores. MSM ameliorated symptoms of pain and improved physical function during the intervention without major adverse events. In another clinical trial, patients with OA of the knee taking MSM daily (1.125 g three times) for 12 weeks showed a decrease in pain and improvement in physical function [55]. In a 12-week trial,[56] treated patients with knee OA with 1.5 g MSM (500 mg three times per day), 1.5 g glucosamine sulfate (GS), MSM plus GS, or placebo for 12 weeks. Significant decreases in the Lequesne Index were reported with MSM, GS, and their combination (P < 0.05). The authors reported a 33% decrease in pain in the MSM group; joint mobility, swelling, global evaluation, and walking time were also improved. The MSM dosage used by [56] was lower than the recommended dosage of MSM for clinical practice. In all of these studies, the improvements were small; therefore, further studies are warranted to establish their clinical significance. For further details on the effects of MSM and DMSO in OA, readers are referred to [57]. Acute and subchronic toxicity studies in rats using a single dose of 2 g/kg and a daily dose of 1.5 g/kg MSM for 90 days showed no adverse events, organ pathology, or mortality [58]. These doses of MSM are considered five times to seven times the maximum dose used in humans [54]. There are no clinical studies on adverse effects, changes in blood chemistry, safety monitoring data, or possible subclinical neurotoxicity symptoms. Side effects of MSM following its oral use are mild and may include digestive upset, headaches, increased blood pressure, increased hepatic enzymes, allergic reactions, and skin rashes.
References:
1. Pavelka K, Trc T, Karpas K, et al. The efficacy and safety of diacerein in the treatment of painful knee osteoarthritis: a randomised, double-blind, placebo controlled multicentre study. Poster presented at: The Annual European Congress of Rheumatology, EULAR; June 8-11, 2005; Vienna, Austria.
2. Brandt KD, Smith G, Kang SY, et al. Effects of diacerhein in an accelerated canine model of osteoarthritis.Osteoarthritis Cartilage 1997;5:438-449
3. Smith GN Jr, Myers SL, Brandt KD, Mickler EA, Albrecht ME. Diacerhein treatment reduces the severity of osteoarthritis in the canine cruciate-deficiency model of osteoarthritis. Arthritis Rheum 1999;42:545-554.
4. Ghosh P, Xu A, Hwa SY, Burkhardt D, Little C. Evaluation of the effects of diacerhein in the sheep model of arthritis [in French]. Rev Prat. 1998;48(17) (suppl):S24-S30.
5. Bendele AM, Bendele RA, Hulman JF, Swann BP. The beneficial effects of diacerein treatment in a guinea pig model of osteoarthritis [in French]. Rev Prat.1996;46:S35-S39.
6. Bendele A, Bendele R, Hulman J, Swann B. A chronic study of the efficacy and toxicity of diacerhein treatment of guinea pigs with osteoarthritis. Abstract presented at: The 2nd OARS International Congress Symposium: Research and Therapeutics in Osteoarthritis: IL-1 Inhibitors; February 4-5, 1995; Nice, France.
7. Mazieres B, Berda L. Effect of diacerhein (art 50) on an experimental postcontusive model of OA [abstract]. Osteoarthr Cartil. 1993;1:47.
8. Douni E, Sfikakis PP, Haralambous S, Fernandes P, Kollias G. Attenuation of inflammatory polyarthritis in TNF transgenic mice by diacerein: comparative analysis with dexamethasone, methotrexate and anti-TNF protocols. Arthritis Res Ther. 2004;6:R65-R72.
9. Dougados M, Nguyen M, Berdah L, et al. Evaluation of the structure-modifying effects of diacerein in hip osteoarthritis: ECHODIAH, a three-year, placebo controlled trial: Evaluation of the Chondromodulating Effect of Diacerein in OA of the Hip. Arthritis Rheum. 2001;44:2539-2547.
10. Hochberg MC, Altman RD, Brandt KD, et al. Guidelines for the medical management of osteoarthritis, II: osteoarthritis of the knee.Arthritis Rheum. 1995;38:1541-1546.
11. Hochberg MC, Altman RD, Brandt KD, et al. Guidelines for the medical management of osteoarthritis, I: osteoarthritis of the hip.Arthritis Rheum. 1995;38:1535-1540.
12. Lequesne M, Brandt K, Bellamy N, et al. Guidelines for testing of slow acting drugs in osteoarthritis. J Rheumatol Suppl. 1994;41:65-73.
13. Pelletier JP, Yaron M, Haraoui B, et al; the Diacerein Study Group. Efficacy and safety of diacerein in osteoarthritis of the knee: a double-blind, placebo controlled trial. Arthritis Rheum. 2000;43:2339-2348.
14. Lequesne M, Berdah L, Gerentes I. Efficacy and tolerance of diacerhein in the treatment of gonarthrosis and coxarthrosis [in French]. Rev Prat. 1998;48(17) (suppl):S31-S35.
15. Tang FL, Wu DH, Lu ZG, Huang F, Zhou YX. The efficacy and safety of diacerein in the treatment of painful osteoarthritis of the knee. Poster presented at: The 11th Asia Pacific League of Associations for Rheumatology (APLAR) Congress, International Convention Center (ICC); September 11-15, 2004; Jeju, Korea
16. Mantia C. A controlled study of the efficacy and tolerability of diacetylrhein in the functional manifestations of osteoarthritis of the hip and the knee: a double blind study versus diclofenac. Unpublished clinical study report; Palermo Hospital, Palmero, Italy; 1987
17. Nguyen M, Dougados M, Berdah L, Amor B. Diacerhein in the treatment of osteoarthritis of the hip. Arthritis Rheum. 1994;37:529-536
18. Louthrenoo W, Nilganuwong S, Aksaranugraha S, et al. The efficacy and safety of diacerein in the treatment of painful osteoarthritis of the knee: a randomised, multicentre, double-blind, piroxicam-controlled, parallel-group, phase III study. Poster Presented at: The 11th Asia Pacific League of Associations for Rheumatology (APLAR) Congress, International Convention Center (ICC); September 11-15, 2004; Jeju, Korea
19. Pietrogrande V, Leonardi M, Pacchioni C. Results of a clinical trial with a new drug, diacerhein in arthrosic patients. Report presented at: The LXXXVI Congress of the Italian National Society of Internal Medicine; September 24, 1985; Sorrento, Italy.
20. Portioli IA. Naproxen-controlled study on the efficacy and tolerability of diacetylrhein in the functional manifestations of osteoarthritis of the knee and hip: a double- blind study versus naproxen. Unpublished clinical study report; Santa Maria Nuova Hospital, Reggio Emilia, Italy; 1987
21. Leeb BF, Bucsi L, Keszthelyi B, et al. Treatment of osteoarthritis of the knee joint. efficacy and tolerance to acemetacin slow release in comparison to celecoxib [in German].Orthopade. 2004;33:1032-1041.
22. Garcia Rodriguez LA, Jick H. Risk of upper gastrointestinal bleeding and perforation associated with individual nonsteroidal anti-inflammatory drugs. Lancet.1994;343:769-772.
23. Brandt KD. Should osteoarthritis be treated with nonsteroidal anti-inflammatory drugs? Rheum Dis Clin North Am. 1993;19:697-712
24. Dieppe P, Cushnaghan J, Jasani MK, McCrae F, Watt IA. Two-year, placebo controlled trial of non-steroidal anti-inflammatory therapy in osteoarthritis of the knee joint. Br J Rheumatol. 1993;32:595-600
25. Juni P, Nartey L, Reichenbach S, et al. Risk of cardiovascular events and rofecoxib: cumulative meta-analysis. Lancet. 2004;364:2021-2029
26. The European Agency for the Evaluation of Medicinal Products. Questions and answers on non-selective NSAIDs: CHMP review of safety of non-selective NSAIDs. London, England: European Agency for the Evaluation of Medicinal Products; 2005. Publication EMEA/250423/2005
27. Food and Drug Administration. Public health advisory non-steroidal anti-inflammatory drug products (NSAIDs). 2006. http://www.fda.gov/cder/drug/advisory/nsaids.htm. Accessed February 1, 2006
28. Setnikar, J., Rovati, L.C., 2001. Absorption, distribution, metabolism and excretion of glucosamine sulfate. A review. Arzneimittelforschung 51, 699–725.
29. Deal, C.L., Moskowitz, R.W., 1999. Nutraceuticals as therapeutic agents in osteoarthritis. The role of glucosamine, chondroitin sulfate, and collagen hydrolysate. Rheum. Dis. Clin. North Am. Huskisson, E.C., 2008. Glucosamine and chondroitin for osteoarthritis. J. Int. Med. Res. 36, 1161–1179 25, 379–395.
30. Hooper, M., 2001. Is glucosamine an effective treatment for osteoarthritic pain? Cleve. Clin. J. Med. 68, 494–495.
31. D’Abusco, A.S., Calamia, V., Cicione, C., et al., 2007. Glucosamine affects intracellular signaling through inhibition of mitogen-activated protein kinase phosphorylation in human chondrocytes. Arthritis Res. Ther. 9, R104.
32. Chan, P.S., Caron, J.P., Rosa, G.J., Orth, M.W., 2005. Glucosamine andchondroitin sulfate regulate gene expression and synthesis of nitric oxide and prostaglandin E2 in articular cartilage explants. Osteoarthritis Cartilage 13, 387–394.
33. Frech, T.M., Clegg, D.O., 2007. The utility of nutraceuticals in the treatment of osteoarthritis. Curr. Rheumatol. Rep. 9, 25–30. Hochberg, M.C., Zhan, M., Langenberg, P., 2008. The rate of decline of joint space width in patients with osteoarthritis of the knee: a systematic review and meta-analysis of randomized placebocontrolled trials of chondroitin sulfate. Curr. Med. Res. Opin. 24, 3029–3035.
34. Jerosch, J., 2011. Effects of glucosamine and chondroitin sulfate on cartilage metabolism in OA: outlook on other nutrient partners especially omega-3 fatty acids. Int. J. Rheumatol. 2011, 1–17.
35. Vangness, C.T., Spiker, W., Erickson, J., 2009. A review of evidencebased medicine for glucosamine and chondroitin sulfate use in knee osteoarthritis. Arthroscopy 25, 86–94.
36. Monfort, J., Pelletier, J.P., Garcia-Giralt, N., Martel-Pelletier, J., 2008. Biochemical basis of the effect of chondroitin sulfate on osteoarthritis articular tissues. Ann. Rheum. Dis. 67, 735–740.
37. Jomphe, C.R., Gabriac, M., Hale, T.M., et al., 2008. Chondroitin sulfate inhibits the nuclear translocation of nuclear factor-kappa B ininterleukin-1beta-stimulated chondrocytes. Basic Clin. Pharmacol. Toxicol. 102, 59–65.
38. Valvason, C., Musacchio, E., Pozzuoli, A., et al., 2008. Influence ofglucosamine sulfate on oxidative stress in human osteoarthritic chondrocytes: effects of HO-I, p22(Phox) and iNOS expression. Rheumatology 47, 31–35.
39. Lopez, V.A., 1982. Double-blind clinical evaluation of the relative efficcacy of ibuprofen and glucosamine sulfate in the management of osteoarthritis of the knee in out-patients. Curr. Med. Res. Opin. 8, 145–149.
40. Muller-Fassbender, H., Bach, G.L., Haase, W., et al., 1994. Glucosamine sulfate compared to ibuprofen in osteoarthritis of the knee. Osteoarthritis Cartilage 2, 61–69
41. Qiu, G.X., Gao, S.N., Giacovelli, G., et al., 1998. Efficacy and safety of glucosamine sulfate versus ibuprofen in patients with knee osteoarthritis. Arzneimittelforschung 48, 469–474.
42. Towheed, T.E., Anastassiades, T.P., Shea, B., et al., 2001. Glucosamine therapy for treating osteoarthritis. Cochrane Database Syst. Rev. CD002946.
43. McAlindon, T., Formica, M., LaValley, M., et al., 2004. Effectiveness of glucosamine for symptoms of knee osteoarthritis: results from an internet-based randomized double-blind controlled trial. Am. J. Med. 117, 643–649.
44. Sawitzke, A.D., Shi, H., Finco, M.F., et al., 2008. The effect of glucosamine and/or chondroitin sulfate on the progression of knee osteoarthritis: a report from the glucosamine/chondroitin arthritis intervention trial. Arthritis Rheum. 58, 3183–3191.
45. Anderson, J.W., Nicolosi, R.J., Borzelleca, J.F., et al., 2005. Glucosamine effects in humans: a review of effects on glucose metabolism, side effects, safety considerations and efficacy. Food Chem. Toxicol. 43, 187–201.
46. Smith, A., Dillon, J., 2009. Acute liver injury associated with the use of herbal preparations containing glucosamine: three case studies.BMJ Case Rep. 1603, 1–8.
47. Hathcock, J.N., Shao, A., 2007. Risk assessment for glucosamine and chondroitin sulfate. Regul. Toxicol. Pharmacol. 47, 78–83.
48. Singh, J.A., Furst, D.E., Bharat, A., et al., 2012. Update of the 2008 American College of Rheumatology recommendation for the use of disease modifying anti-rheumatic drugs and biologic agents in the treatment of rheumatoid arthritis. Arthritis Care Res. 64,625–639.
49. Gupta, R.C., Canerdy, T.D., Lindley, J., et al., 2011. Comparative therapeutic efficacy and safety of type-II collagen (UC-II), glucosamine and chondroitin in arthritic dogs: pain evaluation by ground force plate. J. Anim. Physiol. Anim. Nutr. 96, 770–777.
50. Dechant, J.E., Baxter, G.M., Frisble, D.D., et al., 2005. Effects of glucosamine hydrochloride and chondroitin sulfate, alone and in combination, on normal and interleukin-1 conditioned equine cartilage explants metabolism. Equine Vet. J. 37, 227–231.
51. Breese McCoy, S.J., Bryson, J.C., 2003. High-dose glucosamine associated with polyuria and polydipsia in a dog. J. Am. Vet. Med. Assoc. 222, 431–432.
52. Giaccari, A., Morviducci, L., Zorretta, D., et al., 1995. In vivo effects of glucosamine on insulin secretion and insulin sensitivity in the rat: possible relevance to the maladaptive responses to chronic hyperglycemia. Diabetologia 38, 518–524.
53. Ebisuzaki, K., 2003. Aspirin and methylsulfonylmethane (MSM): a search for common mechanisms, with implications for cancer prevention. Anticancer Res. 23, 453–458.
54. Kim, L.S., Axelrod, L.J., Howard, P., et al., 2006. Efficacy of methylsulfonylmethane (MSM) in osteoarthritis pain of the knee: a pilot clinical trial. Osteoarthritis Cartilage 14, 286–294.
55. Debbi, E.M., Agar, G., Fichman, G., et al., 2011. Efficacy of methylsulfonylmethane supplementation on osteoarthritis of the knee: a randomized controlled study. BMC Complement. Altern. Med. 11, 50.
56. Usha, P., Naidu, M., 2004. Randomized, double-blind, parallel, placebo- controlled study of oral glucosamine, methylsulfonylmethane and their combination in osteoarthritis. Clin. Drug Investig. 24, 353–363.
57. Brien, S., Prescott, P., Bashir, N., et al., 2008. Systematic review of the nutritional supplements dimethyl sulfoxide (DMSO) and methylsulfonylmethane (MSM) in the treatment of osteoarthritis. Osteoarthritis Cartilage 16, 1277–1288.
58. Horvath, K., Noker, P.E., Somfai-Relle, S., et al., 2002. Toxicity of methylsulfonylmethane in rats. Food Chem. Toxicol. 40, 1459–1462.
Marlin
Marlin
(Cholin Bitartrate)
Brand: Marlin
Generic: Cholin Bitartrate
Class: Medical Food Supplement
Route of administration: Oral
Dosage form: Syrup & Tablet
Dose:
Tablet: 1 tablet daily
Syrup: Children: 1 to 2 teaspoon full
Adults: 1 to 2 tablespoon full
Contraindications: Pregnancy: Cat. C
Pack Size: 10 tabs. / 120ml syp.
Price: Rs. 750/-
Supplement facts:
Supplement Amount per serving
Citicoline 500mg
AnMar specs. 300mg
1. Citicoline
Citicoline is the name for cytidine 5′-diphosphocholine (CDP-choline) when this is used as an exogenous sodium salt. In fact, CDP-choline is an endogenous nucleotide naturally found in the body where it is an essential intermediate in the synthesis of the major phospholipid of the cell membranes, phosphatidylcholine (PtdCho). This type of synthesis is called the Kennedy pathway [1].
As a drug, citicoline has been proposed for use in traumatic brain injuries, stroke, vascular dementia, Parkinson’s disease, and brain aging [2] where it has the function of stabilizer of cell membranes and reduces the presence of free radicals [3]. In particular, there is some evidence of a stimulating role of citicoline for the release of dopamine neurotransmitters in the brain [4].
Citicoline, by activating the central cholinergic system, also increases plasma adrenocorticotropic hormone (ACTH) levels and potentiates serum thyrotrophin (TSH) levels. The stimulation of central nicotinic and muscarinic receptors also increases growth hormone (GH) and luteinizing hormone (LH) serum levels [5]. This activity on the cholinergic system is of high therapeutic usefulness in those clinical conditions where alterations of acetylcholine metabolism are considered one of the primary causes of disease [6], eg, Alzheimers Disease (AD).
The biological activity attributed to citicoline has suggested a possible role of citicoline on improving memory [7]. Some clinical studies have given evidence to this hypothesis [8] and there is a proposal for studying citicoline in mild cognitive impairment (MCI) with the aim of confirming both its efficacy in these patients and a possible role as a retardant agent for the cognitive deterioration of the eventual subsequent dementia [9].
1.1. Therapeutic applications of citicoline
1.1.1. Stroke
More than 11 000 people, including patients and volunteers have been studied for evaluating citicoline therapeutic effects. 1372 of these patients were included in studies concerning citicoline efficacy in acute ischemic stroke and pooled together in a meta-analysis of these studies [10]. This analysis showed that citicoline increases the probability of a complete recovery after three months from moderate to severe stroke when it is administered within 24 hours from the event, although the odds ratio of improving with citicoline is only 1.33. Citicoline’s therapeutic action has been attributed to its restoring activity of the PtdCho levels which decrease after a stroke [11].
In stroke patients there seems to be a particular problem in efficiently delivering citicoline at high enough blood levels required to be effective, and to avoid those differences in results evidenced between studies done by intravenous (IV) administration of the drug and oral administration, a more effective preparation for the oral administration is suggested [12].
1.1.2. Vascular dementia
The clinical picture of cognitive and behavioural disorders associated with chronic cerebrovascular disorders (CVD) is much less clear-cut and defined than the picture associated with Alzheimer’s dementia. The definition itself of vascular dementia (VD) has been under discussion for a long time and the heterogeneity of the conditions of patients included in this group is probably higher than the similarities between patients [13].
1.1.3. Correlative studies
When attempts are made to identify relevant relationships between neuroimaging and cognitive patterns, most of the studies have not been able to point out consistent and reliable concordance between these two domains [14] primarily because of the low power of these studies due to the small number of cases [15].
There is evidence of different patterns of cognitive deficits in patients with chronic cerebrovascular disorders when it has been possible to differentiate between those with prevalent signs of altered hippocampal volume from those with diffuse alterations in the grey and white matter [16]. Memory deficits are more relevant in the former group and executive function impairment in the latter group. These findings do not overlap with those that emerge when subcortical dementia is studied as an independent clinical entity characterized by defined quantities of leuakariosis. These neuroimaging findings are commonly associated with deficits in executive function, while memory disorders are considered to be a direct function of cortical impairment [17]. In some instances, this apparent contrast among study results was explained by the different origin of the patients included; eg, when patients from stroke clinics are compared with patients from memory clinics, the specificity of relationship between neuroimaging and functional data is much weaker in the latter group [18]. A proposal has been presented that considers subcortical dementia as a specific form of vascular dementia related to a predominantly small vessel pathogenesis. Mixed dementia is then considered as a nonspecific form of vascular dementia and related to prevalent large infarcts pathogenesis associated with cortical primary atrophy [19]. This model points out the need of considering possible different pathogeneses for different forms of vascular encephalopathy, but does not yet help in identifying specific cognitive patterns of decline associated with them. These issues have largely confounded the studies of citicoline.
1.1.4. Population studies
In studies aimed to identify the relationship between presence of cerebrovascular disorders and prevalence of cognitive disorders in the general population, signs of vascular pathogenesis such as arterial stiffness or generalized atherosclerosis are consistently related to cognitive deterioration which ranges from mild severity to dementia [20].
1.1.5. Specificity of cognitive deficits
The most common way of defining the specificity of cognitive deficits in vascular dementia is based on comparison with AD patients. Cognitive deficits in these two forms of dementia are consistently found to be more severe in AD patients, while the specificity of deficits in vascular dementia is less clear and more difficult to be replicated in diverse studies. Executive function deficits seem to be more prevalent in vascular dementia, while memory deficits are more typical for AD [21].
Another line of research focuses on the identification of specific predictors of developing vascular dementia in groups of patients characterized by those pathologies which are commonly considered as risk factors for cardiovasculopathies such as diabetes and hypertension. The evidence of slight and not evenly distributed signs of cognitive impairment in hypertensive patients [22] and its relationship with the daily temporal distribution of elevated picks of systolic blood pressure (more than with the level of high blood pressure of picks) [23] can be considered as the evidence of a progressive development of a cerebrovascular pathology even before gross anatomical signs of abnormality can be identified in the brain tissues. These findings provide support for the proposal to introduce early therapeutic intervention in patients with mild signs of cerebrovasculopathy or even with risk factors for it [24].
1.1.6. Treatment of cognitive deficits
Attempts to treat symptoms of decline in cerebrovasculopaties have been made since the first years of the last century [25] starting with niacin and continue today. At the beginning of the 1980s, citicoline was used in these clinical areas after having been already in use in treatment for stroke. In most of the clinical studies with citicoline in VD, memory has been the principal end point in the efficacy evaluation. There is a large experimental database of experimental studies on memory and learning performed on aged animals treated with citicoline. Most of these studies have shown that treatment with citicoline ameliorates cognitive deficits but does not necessarily improve normal cognitive functions [26]. The few studies done on memory in aged human subjects with memory disorders, but no dementia, have underlined the extreme variability of positive outcomes depending on the type of patients and the kind of measures used in the studies. Memory is a very complex and multidimensional variable to quantify in humans and, consequently, results from different studies which have in common the assessment of memory are not necessarily homogeneous and comparable if the specific modality of memory evaluation is not taken into account. We have seen that those studies which are trying to identify specific patterns of memory disorders in vascular dementia are not yet able to define a definite and reliable cognitive model of functioning of these patients. As a consequence, it is almost impossible to try to systematically apply a specific memory parameter for the evaluation of treatment efficacy in this clinical area, as it has been possible for AD. There is a possibility that memory disorders might be contaminated by other disorders attributable to executive function including attention. This problem has been examined by looking for methods of evaluating primary memory deficits as distinct from those secondary to other cognitive impairments external to memory per se [27].
Another relevant problem that emerges from a critical analysis of the current and past literature is the relatively poor reliability of single studies performed on small samples of patients with various forms of vascular dementia (a further level of complexity is derived by the different criteria given to these patients in different periods of time). A metanalysis is the best solution available for circumventing these limitations. In the case of citicoline, a meta-analysis for examining the reliability and validity of effects on memory which have been studied in different ways and types of patients in various studies, could fail to confirm the positive results of the single studies once these data are pooled together.
A metanalysis has been performed on the available published and unpublished data obtained from controlled clinical trials done with citicoline. This metanalysis carried out according to the Cochrane Collaboration guidelines is periodically updated in order to include all the studies available [28].
Results of the metanalysis are divided by domains. This allows for a comparative analysis between different areas of assessment; for example, attention and memory. It is possible to verify the homogeneity of results within each domain.
Memory is one of the domains in this analysis and includes results from 884 patients. While studies in this domain include other types of patients as well as cerebrovascular patients, there was no heterogeneity among their results. This indicated that the effect of citicoline on memory was significantly different from the placebo effect, and did not specifically depend on the pathogenesis of the cerebral disorder (effect size 0.19; confidence interval [CI] 95% 0.06, 0.32; p<0.005). In fact, when only cerebrovascular disorders studies were pooled together (for a total of 675 patients), the homogeneity and entity of results was about the same (effect size 0.22; CI 95% 0.07, 0.37; p<0.004).
Within the domain that deals with behavior control and competence (a total of 814 patients), there was a citicoline effect significantly different from placebo and independent from type of measure and pathology examined (effect size −0.26; CI 95% −0.49, −0.04; p<0.004). These results coupled with those of the domain clinical evaluation of improvement concerning a total of 217 patients (effect size determined as the odds ratio of improving under active treatment 8.89; CI 95% 5.19, 15.22; p<0.001) showed that the cognitive effects of citicoline are clearly evident at the behavioral level and can be easily appreciated with a clinical observation of patients irrespective of the functional paradigm used to measure them.
The attention domain, even though based on a substantial number of 790 patients, revealed a large amount of variance because of the large individual differences. These data did not permit an interpretation of how much of the results evidenced by memory measures can be considered as specific of the “true” memory processes or as secondary to an effect on other cognitive components of the cognitive decline.
Finally, the tolerability of citicoline has never constituted a problem whatever the modality of administration or the dosage.
1.2. Conclusions
Treatment in patients with disorders attributed to a cerebrovascular pathogenesis has a long history. Unfortunately, many problems are still unresolved, including the taxonomy of these disorders and a definition of cognitive pattern of decline to be associated to this taxonomy. These intrinsic problems have not helped to develop accepted methods of research and treatment for these patients. Despite these difficulties and elements of confusion between different clinical studies performed at different periods of time, citicoline has emerged as a valid treatment for patients with chronic cerebrovascular disorders or with memory problems
References:
1. Fernandez-Murray JP, McMaster CR. Glycerophosphocholine catabolism as a new route for choline formation for phosphatidylcholine synthesis by Kennedy pathway. J Biol Chem. 2005;46:38290–6.[PubMed]
2. Blount PJ, Nguyen CD, McDeavitt JT. Clinical use of cholinomimetic agents: a review. J Head Trauma Rehabil. 2002;17:314–21. [PubMed]
3. Zweifler RM. Membrane stabilizer: citicoline. Curr Med Res Opin. 2002;18(Suppl 2):14–17.[PubMed]
4. Fonlupt P, Martinet M, Pacheco H. Effect of CDP-choline on dopamine metabolism in central nervous system. In: Zappia V, Kennedy EP, Nilsson BI, et al., editors. Novel biochemical, pharmacological, and clinical aspects of CDP-choline. New York: Elsevier; Sci: 1985. pp. 169–77.
5. Cavum S, Savci V. CDP.choline increases plasma ACTH and potentiates the stimulated release of GH, TSH, and LH: the cholinergic involvement. Fundam Clin Pharmacol. 2004;18:513–23.[PubMed]
6. Shen ZX. Brain cholinesterases: III. Future perspectives of AD research and clinical practice. Med Hypotheses. 2004;63:298–307. [PubMed]
7. McDaniel MA, Maier SF, Einstein GO. “Brain-specific” nutrients: a memory cure? Nutrition. 2003;19:957–75. [PubMed]
8. Agnoli A, Bruno G, Fioravanti M. Therapeutic approach to senile memory impairment: a double-blind clinical trial with CDP-choline. In: Wurtman RJ, Corkin S, Growdon JH, et al., editors. Alzheimer’s disease: proceedings of the fifth meeting of the International Study Group in the Pharmacology of Memory Disorders Associated with Aging. Boston: Birkhauser; 1986. pp. 649–54.
9. Abad-Santos F, Novalbos-Reina J, Gallego-Sandin S, et al. Tratamiento del deterioro cognitivo lieve: utilidad de la citicolina. Rev Neurol. 2002;35:675–82. [PubMed]
10. Davalos A, Castillo J, Alvarez-Sabin J, et al. Oral citicoline in acute ischemic strike: an individual patient data pooling analysis of clinical trials. Stroke. 2002;33:2850–7. [PubMed]
11. Rao AM, Hatcher JF, Larsen EC, et al. CDP-choline significantly restores the phosphatidylcholine levels by differentially affecting phospholipase A2 and CTP-phosphocholine cytidylyltransferase after stroke. J Biol Chem. 2006;281:6718–25. [PubMed]
12. Rao AM, Hatcher JF. Cytidine-5’-Diphosphocholine (CDP-choline) in stroke and other CNS disorders. Neurochem Res. 2005;30:15–23. [PMC free article] [PubMed]
13. Wallin A, Milos V, Sjogren M, et al. Classification and subtypes of vascular dementia. Int Psychogeriatr. 2003;15(suppl 1):27–37. [PubMed]
14. Paul RH, Cohen RA, Moser DJ, et al. Clinical correlates of cognitive decline in vascular dementia. Cogn Behav Neurol. 2003;16:40–6. [PubMed]
15. Cohen RA, Browndyke JN, Moser DJ, et al. Long-term citicoline (Cytidine Diphosphate Choline) use in patients with vascular dementia: neuroimaging and neuropsychological outcomes. Cerebrovasc Dis. 2003;16:199–204. [PubMed]
16. Mungas D, Harvey D, Reed BR, et al. Longitudinal volumetric MRI change and rate of cognitive decline. Neurology. 2005;65:565–71. [PMC free article] [PubMed]
17. Price CC, Jefferson AL, Merino JG, et al. Subcortical vascular dementia: integrating neuropsychological and neuroradiologic data. Neurology. 2005;65:376–82. [PMC free article][PubMed]
18. Rockwood K, Black SE, Song X, et al. Clinical and radiological subtypes of vascular cognitive impairment in a clinic-based cohort study. J Neurol Sci. 2006;240:7–14. [PubMed]
19. Jellinger KA. Pathology and pathophysiology of vascular cognitive impairment. A critical update. Panminerva Med. 2004;46:217–26. [PubMed]
20. Hanon O, Haulon S, Lenoir H, et al. Relationship between arterial stiffness and cognitive function in elderly subjects with complaints of memory loss. Stroke. 2005;36:2193–7. [PubMed]
21. Traykov L, Baudic S, Raoux N, et al. Patterns of memory and perseverative behavior discriminate early Alzheimer’s disease from subcortical vascular dementia. J Neurol Sci. 2005:229–230. 75–9.[PubMed]
22. Fioravanti M, Agazzani D, D’Ilario D, et al. Relationship between hypertension and early indicators of cognitive decline. Dementia. 1991;2:51–6.
23. Fioravanti M, Nacca D, Golfieri B, et al. The relevance of continuous blood pressure monitoring in examining the relationship of memory efficiency with blood pressure characteristics. Physiol Behav. 1996;59:1077–84. [PubMed]
24. Schindler RJ. Dementia with cerebrovascular disease: the benefits of early treatment. Eur J Neurol. 2005;12(suppl 3):17–21. [PubMed]
25. Fioravanti M. History of symptomatic therapy in vascular dementia. Int Psychogeriatr. 2003;15(suppl 1):177–81. [PubMed]
26. Conant R, Schauss AG. Therapeutic applications of citicoline for stroke and cognitive dysfunction in the elderly: a review of the literature. Altern Med Rev. 2004;9:17–31. [PubMed]
27. Fioravanti M, Di Cesare F. Memory improvements and pharmacological treatment: a method to distinguish direct effects on memory from secondary effects due to attention improvement. Int Psychogeriatr. 1992;4:119–26. [PubMed]
28. Fioravanti M, Yanagi M. Cytidinediphosphocholine (CDP-choline) for cognitive and behavioural disturbances associated with chronic cerebral disorders in the elderly. Cochrane Database Syst Rev. 2005;18:CD000269. [PubMed]
StoneMar Plus
Sed ut perspiciatis unde omnis iste natus error sit voluptatem accusantium doloremque laudantium,
[…]
Tab LMC
Tab LMC
(L-methyl folate + Methylcobalamin +N-Acetylcycteine)
Brand: LMC
Generic: L-methyl folate + Methylcobalamin +N-Acetylcycteine
Class: Medical Food Supplement
Route of administration: Oral
Dosage form: Tablet
Dose: 1 tablet twice a day
Pregnancy: Safe
Pack Size: 30 tabs.
1. L-METHYLFOLATE
Folic acid and Methyl folate both are from Vitamin B9, but folic acid is man-made (synthetic) where methylfolate is obtained from natural source. L-methylfolate, the active form of folate is essential for biological processes throughout the human system involving methylation or one-carbon transfer. Without it there can be a significant breakdown of the human body’s ability to produce healthy DNA, have healthy neurotransmitter production, and optimal cardiovascular, hormonal, cellular, liver and reproductive functions. Folate deficiency is tied to a host of conditions. Because L-methylfolate is required for the synthesis of the neurotransmitters serotonin, dopamine, and norepinehrine, low levels make it less likely that people will respond well to products that support a healthy mood. L-Methylfolate is also the only form of folate able to cross the blood-brain barrier.
A decline of folate levels in the cerebral spinal fluid has been associated with reduced cognitive function. L-Methylfolate is essential for the conversion of homocysteine to methionine, supporting cardio and vascular health by lowering homocysteine levels. Since L-methylfolate participates in DNA synthesis and cell division, requirements are increased during pregnancy and periods of rapid growth. Although folate occurs in naturally in some foods, and folic acid is used to fortify others, approximately 60 percent of the population in the United States have genetic variations that make them unable to utilize these nutrients. These variations do not allow the MTHFR enzyme to function properly so methylation of folate does not occur. Without this activation step, folate cannot be used by the body, resulting in negative health side effects and the possible inability to maintain a healthy mood. This deficiency usually occurs because folic acid, the most common form of folate in supplements and fortified foods, must be converted to its active forms to be used by the body. The enzyme needed to convert folate to its active form, methylfolate, is called methylenetetrahydrofolate reductase (MTHFR). The problem lies in the fact that almost half of all people have genetic variations that reduce the ability of the MTHFR enzyme to make this conversion. Folate deficiencies are tied to a host of conditions.
For people with MTHFR gene variations, supplementing with the already activated form of folate is far more effective in providing this form of folate than introducing the pre-converted form to the body through typical folate supplements. L-Methylfolate supplements provide theactive form of folate naturally present in the bodyand available for biological action.
This image illustrates the folic acid activation cycle. The other forms of vitamin B9 are folic acid and folate. Folate is often called the natural form of vitamin B9, but it actually refers to a family of different compounds that occur naturally in some vegetables. Folic acid is the supplemental or “synthetic” form of vitamin B9 and is typically the first line of treatment for folate deficiency and related health conditions, such as high homocysteine and neural tube defects. Any folic acid we ingest must be metabolized (converted) into Dihydrofolate (DHF), Tetrahydrofolate (THF), and then finally into L-methylfolate to be used in the body. Once in this form it can be transported into cells, tissues and even across the blood-brain barrier. But issues can arise during metabolism for those with an enzyme issue, such as an MTHFR mutation.
1.1. SIGNS AND SYMPTOMS OF AN MTHF DEFICIENCY
1.1.1.Low Brain Chemistry Production
L-Methylfolate is essential for maintaining healthy brain chemistry and is responsible for the normal production of the neurotransmitters serotonin, melatonin, dopamine, epinephrine, and norepinephrine. These neurotransmitters are involved in a number of functions including short-term memory, concentration, sleep, motor control, hormone control, mood stability, motivation, and appetite control. Low folate levels have been associated with mood disorders and supplemental folate might be recommended as first line therapy.
1.1.2. Homocysteine
Genetic polymorphisms in methylenetetrahydrofolate reductase (MTHFR) have been proven to be the common hereditary factors of high-level homocysteine. Supporting normal homocysteine levels already in a normal range is important to good health. L-Methylfolate supplements address elevated homocysteine levels in the blood and improve peripheral blood flow in the arteries by increasing nitric oxide production in the vascular endothelium. This mode of action supports heart health.
1.1.3. Peripheral Neuropathy
L-Methylfolate supplements can help promote healthy peripheral nerve function by supporting normal homocysteine levels and increasing nitric oxide production.
1.1.4. Uric Acid
Gout is a condition characterized by the deposition of monosodium urate crystals in the joints or soft tissue. Acute gout most commonly affects the first metatarsal joint of the foot, but other joints are also commonly involved. Supplementing with L-methylfolate can aide in the normal metabolism of uric acid.
1.1.5.Neural Tube Defects
Women who consume healthful diets with adequate folate throughout their childbearing years may reduce their risk of having a child with a birth defect of the brain or spinal cord. Sources of folate include fruits, vegetables, whole grain products, fortified cereals, and dietary supplements. Supplementing with folate significantly reduces the occurrence of neural tube defects. However, a study involving a group of women of childbearing age showed that supplementing with L-methylfolate results in a higher red blood cell folate level than supplementing with folic acid.
2. N-ACETYL CYSTEINE
Acetylcysteine is the N-acetyl derivative of the amino acid L-cysteine, and is a precursor in the formation of the antioxidant glutathione in the body. The thiol (sulfhydryl) group confers antioxidant effects and is able to reduce free radicals.
2.1. BENEFITS OF N-ACETLY CYSTEINE
2.1.1. Glutaminergic Neurotransmission
The cystine–glutamate antiporter(also called exchanger or counter-transporter) on astrocytes is known to mediate synaptic levels of glutmate, and takes up cystine (the dimer of cysteine) in exchange for glutamate release, which includes inhibitory metabotropic glutamate receptors (known as group II) on glutaminergic nerve terminals and reduced further synaptic release of glutamate. It is thought that provision of additional cysteine, via N-acetylcysteine supplementation, can provide more substrate for this reaction to reduce glutaminergic stimulation.
This antiporter may also be downregulated and its function disturbed with usage of drugs, which is involved in the pathology of addiction;N-acetylcysteine may attenuate the reduction seen with drug administrationand disrupt the transporters autoregulation.
It is thought that additional cysteine in the brain can reduce glutaminergic transmission by reducing the amount of neuronal glutamate release into the synapse (secondary to aiding its release from astrocytes).
In brain injury or disease, they often work in reverse, and excess glutamate can accumulate outside cells. This process causes calcium ions to enter cells via NMDA receptor channels, leading to neuronal damage and eventual cell death, and is called excitotoxicity. The mechanisms of cell death include
• Damage to mitochondria from excessively high intracellular Ca2+
• Increased intracellular nitric oxide concentrations[7]
• Glu/Ca2+-mediated promotion of transcription factors for pro-apoptotic genes, or downregulation of transcription factors for anti-apoptotic genes
Excitotoxicity due to excessive glutamate release and impaired uptake occurs as part of the ischemic cascade and is associated with stroke, autism, some forms of intellectual disability, and diseases such as amyotrophic lateral sclerosis, lathyrism, and Alzheimer's disease. In contrast, decreased glutamate release is observed under conditions of classical phenylketonuria leading to developmental disruption of glutamate receptor expression.
Glutamic acid has been implicated in epileptic seizures. Microinjection of glutamic acid into neurons produces spontaneous depolarisations around one second apart, and this firing pattern is similar to what is known as paroxysmal depolarizing shift in epileptic attacks. This change in the resting membrane potential at seizure foci could cause spontaneous opening of voltage-activated calcium channels, leading to glutamic acid release and further depolarization.
2.2. N-acetyl cysteine in Pregnancy
N-acetyl cysteine is usually safe for pregnant women to take, but ask your doctor if you are pregnant or trying to get pregnant before taking this supplement. Taking this supplement may help you get pregnant and prevent a miscarriage under certain circumstances.
2.2.1. Increasing Ovulation
People who have polycystic ovary disease may experience an improvement in fertility when taking N-acetyl cysteine along with fertility drugs. Polycystic ovary disease is the most common hormonal disorder among women of reproductive age that the cause is unknown, according to the Mayo Clinic. It causes a lack of ovulation and therefore a difficulty getting pregnant. A 2007 study published in the "ActaObstetricia et GynecologicaScandinavica" journal found that N-acetyl cysteine induced or augmented ovulation in polycystic ovary patients. Participants took 50 mg of the fertility drug clomiphene citrate twice a day alone or combined with 1,200 mg daily of N-acetyl cysteine for five days. Medication began on the third day of their menstrual cycle. The study was entitled, "N-Acetyl Cysteine and Clomiphene Citrate for Induction of Ovulation in Polycystic Ovary Syndrome: A Cross-over Trial."
2.2.2. Decreasing Miscarriages
Taking N-acetyl cysteine reduces the possibility of pregnant women miscarrying. N-acetyl cysteine may suppress oxidative stress, according to a 2008 study published in the "Reproductive BioMedicine Online" journal. Oxidative stress happens when free radicals attack and destroy healthy brain cells. This may begin a cascade of changes in the body that causes a miscarriage. Humans convert N-acetyl cysteine into the antioxidant, glutathione. Antioxidants fight free radicals. The study found that N-acetyl cysteine and folic acid supplements significantly increased the take-home baby rate of participants compared to taking folic acid alone. The name of the study is "N-acetyl Cysteine for Treatment of Recurrent Unexplained Pregnancy Loss.
3. METHYLCOBALAMIN
Methylcobalamin circulates through the bloodstream and is one of two naturally occurring coenzyme forms of vitamin B12 that the body utilizes. It differs from typical B12 in that it has an additional methyl group, and contains metal-alkyl bonds. Adenosylcobalamin is the other form sometimes used in the dietary supplement industry. The body needs B12 to convert homocysteine to methionine, protect DNA and RNA, support energy, protect nerve and brain cells, stimulate serotonin production, contribute to red blood cell formation, support immune function, and maintain a positive mood.
Hydroxocobalamin is the main B12 form found in meat, fish, and dairy products. Cyanocobalamin is a common but synthetic B12 that is often added to foods, and the body must use precious energy to convert this form into methylcobalamin. This B12 form has a higher stability and bioavailability and doesn’t require conversion. These next 4 facts will explain the benefits of methylcobalamin.
3.1. BENEFITS OF METHYLCOBALAMIN
3.1.1. Support Brain and Nerve Health
Methylcobalamin creates two major effects in the brain which are responsible for many of its neuro-protective and longevity (long life) enhancing benefits.
First, the compound eliminates excessive levels of homocysteine in the brain. A high level of homocysteine in the blood (hyperhomocysteinemia) makes a person more prone to endothelial cell injury, which leads to inflammation in the blood vessels, which in turn may lead to atherogenesis, which can result in ischemic injury. This substance is often cited as the leading cause of vascular degeneration in the brain.
Next, Methylcobalamin improves the health and function of nerves in the brain. It can rebuild damaged nerves and contributes to the maintenance of function that is normally decayed during the aging process.
For people with autoimmune disorders, blood cell conditions like anemia can lead to immunity issues and cognitive impairment. Methyl B12 aids this problem by allowing intrinsic factor to bind appropriately in the intestines.It also raises the available amount of SAMe (S adenosylmethionine), the important compound which lifts mood and works as a tricyclic antidepressant.Higher amounts of methylcobalamin are necessary to correct neurological defects and protect against aging. Published studies show that high doses of methylcobalamin are needed to regenerate neurons as well as the myelin sheath that protects nerve axons and peripheral nerves.
Few substances have been shown to regenerate nerves in humans with peripheral neuropathies. However, a study in the Journal of Neurological Science postulated that methylcobalamin could increase protein synthesis and help regenerate nerves. The scientists showed that very high doses of methylcobalamin produce nerve regeneration in laboratory rats.
3.1.2. Increases Available Physical Energy
Every chemical reaction in the body expends energy. When the body needs to convert a vitamin into a form the body can use, this reaction also requires energy. For example, supplementing with the cyanocobalamin form of B12 requires the body to expend energy that removes the cyanide molecule and replaces it with a methyl group. Taking methylcobalamin as a B12 supplement eliminates the need for this extra chemical reaction.
3.1.3. Encourages Detox
Methyl groups activate hundreds of chemical reactions throughout the body. One such process includes triggering detox reactions. This includes the removal of heavy metals, environmental toxins, and waste products.
3.1.4. Neutralizes Homocysteine
High homocysteine levels is undesirable and known to be an indicator of heart disease and stroke risk. Free homocysteine in the blood causes sclerosis of the arteries, putting strain on the vascular system and the heart. One of the primary reactions of methylcobalamin is to convert homocysteine to methionine, reducing the potential for damage. Outcomes from this reaction include the formation of cysteine, a precursor to the super-antioxidant glutathione. Methionine also contributes to the formation of adenosylcobalamin, the other form of B12 used by the body in mitochondrial energy creation, the foundation for all human energy.
UD-Care
Cap UD-Care
(Cranberry + Probiotics)
Brand: UD-Care
Generic: Cranberry + Probiotics
Class: Medical Food Supplement
Route of administration: Oral
Dosage form: Capsule
Dose: 1 capsule daily
Contraindications: Pregnancy: Cat. C
Pack Size: 10 tabs.
Price: Rs. 650/-
Supplement facts:
Total Probiotic Blend 5 billion
Lactobacillus acidophilus 1.5 billion CFU
Lactobacillus rhamnosus 1.5 billion CFU
Lactobacillus casei 750 million CFU
Lactobacillus fermentum 500 million CFU
Bifidobacterium longum 750 million CFU
Cranberry 36:1 (Vaccinium macrocar-pon) Powdered fruit extract 200 mg
1. Cranberry Juice
Cranberry is produced from the berry fruit of a North American evergreen shrub. Cranberry is acidic and can interfere with unwanted bacteria in the urinary tract. Cranberry is also believed to act as a diuretic ("water pill").Cranberry (as juice or in capsules) has been used in alternative medicine as a possibly effective aid in preventing symptoms such as pain or burning with urination. Cranberry will not treat the bacteria that causes a bladder infection [1].
Infection occurs when bacterial adhesins located on its outer cell wall interact with mucosal glycoproteins and epithelial mucins. This mechanism has been recently exploited to target the adhesive process of H. pylori. Compounds such as polysaccharides are found to interact with the bacterial adhesins before adhesinmucin, thus avoiding the infection process [2]. Because adhesion to epithelial cells is the most essential step of the infectious process for almost all bacterial pathogens, many efforts have been aimed at developing antiadhesin therapy. Sialyllactose (NeuAc[K2-3] Gal[L1-4]Glc), an inhibitor of the sialic acid-specific adhesin of H. pylori, was found to reduce the load of bacteria in monkeys. Cranberry juice has also been found to be useful in treating H. pylori infection via inhibition of sialic acid-specific adhesion of H. pylori to human gastric mucus and to human erythrocytes [3]. Similarly, root extracts of Pelargonium sidoides DC (Geraniaceae), which contain a polysaccharide fraction, EPs 7630, with antiadhesive activity against H. pylori, was found to be useful for treating acute respiratory infections [4]. Glycyrrhzia glabra L. root is a rich source of polysaccharides composed of arabinose, galactose, glucose, and glucuronic acid and is able to interact with the outer membrane surface adhesins of H. pylori, avoiding its adhesion to mucus [4].
2. PROBIOTICS:
2.1. Introduction
The term “probiotic” is derived from the Greek and means “for life”; however, it has had several different meaning over the years. The history of probiotics began with the consumption of fermented foods by Greeks and Romans. Probably the first foods that contained living microorganisms were the fermented milks that are recorded in the Old Testament (Genesis 18:8), which reported that “Abraham owed his longevity to the consumption of sour milk.” The consumption of fermented milks in many different forms has continued until the present day. The beneficial effects of yogurt were found scientifically in the early of the twentieth century. Ilya Ilyich Metchnikoff (1908, 1910), a Nobel Prize winner working at the Pasteur Institute in Paris, France, was the first who proposed the beneficial effect of fermented milk on human health and regarded the microflora of the lower gut as having an adverse effect on the health of the human adult. He put forward the theory that the effect of detrimental microbes in the intestinal tract could be alleviated by the ingestion of beneficial microbes. Metchnikoff also postulated that the longevity of the Bulgar mountain people was attributable to the consumption of the yogurt produced by fermentation with Lactobacillus bulgaricus (previously called the Bulgarian bacillus). It should be emphasized that Metchnikoff was not concerned with sour milk but rather what we now call yogurt; subsequently, when pure cultures became available he advocated the use of milk fermented with a single strain of Lactobacillus. So, probiotics and the phenomenon of probiosis were discovered by Metchnikoff. Probiosis can be defined as the positive effect of consumption of fermented dairy products with cultures of lactic acid bacteria (LAB) on the equilibrium of intestinal microflora. It is presently known that viable LAB implantation in the digestive tract act directly on the composition of the microbial population. Metchnikoff’s work can be regarded as the birth of probiotics, that is, microbes ingested with the aim of promoting good health. The habit was given added support by the publication of the book “The Bacillus of Long Life” in 1911, where the author, Loudon Douglas, reiterated the connection between fermented milk and longevity [6]. The term probiotic, as is used today, was first used in 1974 by Parker, who defined probiotics “as substances and organisms that contribute to intestinal microbial balance.” During this period there grew a realization that the gut microflora was involved in protection of the host against disease. These studies reinforced the view that not all bacteria were having adverse effects on the host and that there was a population of bacteria in the gut necessary for the continuing positive health and wellbeing of the host by improving its intestinal microbial balance [7]. This definition was broadened by [8], who included in the definition the following: “a viable mono- or mixed culture of microorganisms that, applied to animal or man, beneficially affects the host by improving the properties of the indigenous microflora.” This implies that the term probiotic is restricted to products that: (i) contain live microorganisms (such as freeze-dried cells or in a fermented product); and (ii) improve the health status of humans or animals and exert their effects in the mouth or gastrointestinal tract (GIT) (aerosol) or in the urogenital tract (by local application). Probiotics are defined in human nutrition as “live microorganisms that can provide benefits to human health when administered in adequate amounts that confer a beneficial health effect on the host” [9]. There is an increasing amount of evidence indicating health benefits by their consumption [10]. Temporary colonizing of the gut with an appropriate probiotic strain not only promotes the state of eubiosis (favorable balance of the gut flora) but also may have a favorable immunomodulatory effect [11]. The probiotic products can contain one or several species of probiotic bacteria; however, there is still a significant way to go before the association of specific probiotic health benefits with specific strains of bacteria. Probiotics are commonly defined as “viable microorganisms (yeast and LAB) that exhibit a beneficial effect on the health of the host when they are ingested, although its health benefits are strain-specific and not species-specific or genus-specific.” Many physiological responses and health-promoting effects in the consumer attributed to probiotic microorganisms are related, among others, to the GIT, and show the ability to survive through the upper GIT and the capability of surviving and growing in the intestine (acid and bile resistant) in a strain-specific manner safe for human consumption.
They produce antimicrobial substances like bacteriocins (nisin and pediocin), can adhere to human intestinal cell lines, and colonize the intestine. Probiotics produce short-chain fatty acids (SCFA) that lower the pH, thus favoring the growth of harmless microorganisms, including low-molecular-weight carboxylic acids with six to eight carbon atoms (i.e., acetate, propionate, butyrate). With this acidic pH, probiotics can easily penetrate the bacterial cell. Probiotics are live microorganisms, generally bacteria but also yeast, that interact with the gut microflora and host when ingested in a sufficient amount. They have a positive effect on the health of an individual beyond the nutritional ones commonly known. These bacteria can help to maintain internal microbial balance and defend against harmful bacteria; three mechanisms of promoting human health has been described: (i) providing endproducts of anaerobic fermentation of carbohydrates such as organic acids that can be absorbed by the host, these end-products being able to influence human mood, energy level, and even cognitive abilities; (ii) successfully competing with pathogens; and (iii) stimulating host immune responses by producing specific polysaccharides [12]. Probiotics for consumption should not cause disease in humans, should be completely nonpathogenic, and should not be able to evolve into pathogenic variants. Probiotics are readily available to consumers and are commonly found as food probiotics (e.g., yogurts, cheeses, milk-based beverages, fermented fish, meats, and vegetables, among others) and as food supplement probiotics (e.g., tablets, capsules, pills, powders, liquid concentrates in vials, and soft gels, among others) [13]. Although probiotics can technically be any type of beneficial microorganism, certain bacteria are more commonly found on the food market.
Currently, the most widely used food-grade probiotics in the majority of countries include LAB (LAB), from the genera Lactobacillus, and Bifidobacteria, with some strains of Enterococcus and Saccharomyces species being among the exceptions. LAB are Gram-positive anaerobic aero-tolerant nonspore-forming rods and cocci that are indigenous inhabitants of the human GIT, vagina, and human skin.
Lactobacillus acidophilus and Bifidobacterium represent two distinct phyla of bacteria, the Firmicutes and Actinobacteria; even though they are genetically quite distinct, they have many phenotypic similarities (e.g., producers of lactic acid that can easily be added to traditionally fermented foods, such as cheeses, yogurts, and other dairy products). For this reason, Bifidobacteria are often included in the LAB, but because the LAB are a taxonomic group of genetically similar bacteria, the Bifidobacteria cannot be included from a taxonomic point of view. Both are inhabitants of GI flora of humans and animals, with the Lactobacilli believed to be important for small intestine functionality and Bifidobacteria for colon functionality [13].
2.2. Probiotics and their sources
The probiotic bacteria generally belong to the Lactobacillus and Bifidobacterium genera. However, other bacteria and some yeast also have probiotic properties. Common bacteria include the following [14]:
● Lactic acid bacteria (LAB): Genus: Lactobacilli spp.; Species: Lactobacillus acidophilus, L. amylovorus, L. brevis, L. bulgaricus, L. casei, L. cellobiosus, L. crispatus, L. curvatus, L. delbrueckii spp. bulgaris, L. fermentum, L. gallinarum, L. helveticus, L. johnsonii, L. lactis, L. paracasei, L. plantarum, L. reuteri, L. rhamnosus;
Genus: Streptococcus spp. Species: Streptococcus salivaris spp. thermophiles;
Genus: Lactococcus ssp., Species: L. lactis cremoris; Genus: Leuconostoc, Species: Lc. mesenteroides; and
Genus: Pediococcus spp., Species: P. pentosaceus, P. acidilactici. Bifidobacteria: Genus: Bifidobacterium spp., Species: B. adolescentis, B. animalis, B. bifidum, B. breve, B. essensis, B. infantis, B. laterosporum, B. thermophilum, B. longum.
● Propionibacteria: Genus: Propionibacterium spp., Species: P. acidipropionici, P. freudenreichii, P. jensenii,
P. thoenii.
● Enterobacteria: Genus: Enterococcus spp., Species: E. fecalis, E. faecium.
● Sporulated bacteria: Genus: Bacillus spp., Species: B. alcolophilus, B. cereus, B. clausii, B. coagulans, B. subtilis.
● Other bacteria: Genus: Escherichia coli, Species: E. coli; Genus: Sporolactobacillus spp. Species: S. inulinus.
● Yeasts: Genus: Saccharomyces spp., Species: S. cerevisae (boulardii); that isolated from litchi fruit in Indonesia have also been accepted and used as probiotics.
To be considered as probiotics, the different strains should be normal inhabitants of a healthy intestinal tract, survive the upper digestive tract, be capable of surviving and growing in the intestine (acid and bile resistant), be safe for human consumption, produce antimicrobial substances (i.e., bacteriocins), and have the ability of adherence to human intestinal cell lines and colonization. [15] established the criteria for microorganisms to be included in the probiotic group as follows: (i) surviving on passing through the GIT at low pH and on contact with bile; (ii) adhesion to intestinal epithelial cells; (iii) stabilization of the intestinal microflora; (iv) nonpathogenic; (v) survival in foodstuffs and possibility for production of pharmacopeia lyophilized preparations; (vi) fast multiplication, with either permanent or temporary colonization of the GIT; and (vii) generic specificity of probiotics.
2.3. Mechanism of Action of Probiotics
The mechanistic approach to probiotics first established than many GIT dysfunctions are based on disturbances or imbalances of intestinal microflora. The productions of inhibitory substances by probiotics such as organic acids, hydrogen peroxide, and bacteriocins are of much interest. However, the mechanisms whereby Lactobacilli function as anti-infective defenses are still not fully understood [13]. The actions of microorganisms are useful to assist the GIT by breaking down sugars and carbohydrates to promote good digestion, boost the immune system, maintain proper intestinal pH, and successfully compete with pathogens. Among the expectations for probiotics, many strains have been shown to modulate the intestinal microflora and to prevent the duration and symptoms of rotavirus-induced diarrhea; probiotic bacteria also reinforce the intestinal wall by crowding out pathogenic microorganisms, therefore helping to prevent their attachment to the human gut, where they have been shown to be safe. The consumption of probiotics has been shown to influence various aspects of the innate nonspecific immune system like promotion of mucin production, inhibition of pathogenic bacteria, decrease in gut permeability, macrophage activation, and phagocytic capacity, and natural killer (NK) cell activity. Regarding the adaptive immune system, the effects observed are an increase in the production of antibodies (IgA, IgM, and IgG), and also an influence in the arrangement of both branches of the immune system by the production of cytokines and other regulatory elements. Hydrogen peroxide is produced by many species of Lactobacillus because antimicrobial agents are oxygen dependent. Because Lactobacillus does not produce catalase, the hydrogen peroxide produced cannot be degraded and acts as an oxidant by forming free radicals. Bacteriocins are proteinaceous antimicrobial substances that are sometimes associated with lipids and carbohydrates. Bacteriocins have demonstrated inhibitory actions in both Gram-positive and Gram-negative bacteria. Probiotics usually modify the resident microflora. It is known that probiotic bacteria alter the physical environment so that the pathogenic bacteria cannot survive. Probiotic bacteria act in two ways.
First, they compete with pathogenic bacteria for food and energy sources.
Second, they produce an inhibitory substance that impedes the growth of the pathogen by consuming the nutrients that pathogens need. Probiotics and pathogenic bacteria are in competition; probiotics inhibit the pathogens by adhering to the intestinal epithelial surfaces and blocking the adhesion sites [13]. The second generation of probiotics are genetically modified microorganisms that provide the host with some necessary components (e.g., production of immunomodulators, such as interleukines, or Helicobacter pylori and rotavirus antigens) [16]. Probiotics have several putative mechanisms such as modulation of immune or sensory-motor function, enhancement of mucosal barrier function, and antipathogen effects. The epithelial barrier consists of a thick mucus layer containing immunoglobulins, mainly IgA and antimicrobial components; their dynamic functional role is to regulate permeability between cells. Intestinal mucosal barrier function is formed by a common mucosal immune system that provides communication between the different mucosal surfaces of the body. In the future, a greater understanding of probiotic-specific mechanisms could allow for precise selection of a particular probiotic strain to target a patient’s specific pathogen defect and clinical problem [17].
2.4. PROBIOTICS USED IN FOOD OR AS MEDICINES
Most probiotics are marketed as foodstuffs or medicines. Lactobacillus, Leuconostoc, and Pediococcus species have been used extensively in food processing throughout human history, and ingestion of foods containing live bacteria, dead bacteria, and metabolites of these microorganisms has taken place for a long time [18]. Currently, the most widely used probiotics include Lactobacilli, Bifidobacteria, and some nonpathogenic strains, mostly of human origin, that confer health benefits to the host and enable prevention of or improvement in some diseases when administered in adequate amounts. An important fact is that probiotics must retain their viability during the storage, manufacturing process, and transit through the stomach and small intestine. Prior to being categorized as probiotics, organisms need to follow a process of testing, including strain testing, identification by genotype and phenotype, functionalized characterization and safety assessment testing, and double-blind, placebo-controlled human trials to verify their health benefits; the guidelines for the evaluation of probiotics in food have been proposed [9].
Most probiotic foods contain Lactobacilli and/ or Bifidobacteria. Microorganisms used as probiotics in animals are mainly bacterial strains of members of the heterogeneous group of LAB: Lactobacilli (L. acidophilus, L. casei, L. plantarum, L. reuteri, L. rhamnosus, L. salivarus), Bifidobacteria (B. breve, B. longum, B. lactis), Bacillus (B. subtilis), and Enterocococcus (E. faecium), among others. The yeast Saccharomyces boulardi is also used as a human probiotic, although it is delivered in capsules or powders rather than by food. It is noticed that Bacillus and Lactobacillus differ in many characteristics and that the Bacillus and the yeasts are not usual components of the gut microflora. It has been shown that most Lactobacillus species (L. acidophilus, L. rhamnosus, and L. reuteri) have no pathogenicity or acute oral toxicity in animals [13]. The use of probiotics in medical practice is important. The effects and applications of probiotics in various infectious diseases have been summarized by [19], who categorized into four groups the clinical applications for probiotics by level of evidence of efficacy. The first group includes the application to cases of acute/antibiotic-associated gastroenteritis, for which benefits of probiotics are well proven. The second group includes allergic reactions, specifically atopic dermatitis I, for which there is substantial evidence of efficacy. The third group includes applications that have shown promise (e.g., childhood respiratory infections, dental caries, inflammatory bowel disease, combating nasal pathogens, and the prevention of relapsing Clostridium difficile-induced gastroenteritis). The fourth group covers potential future applications for rheumatoid arthritis, irritable bowel syndrome, cancer, alcohol-induced liver disease, diabetes, and graft-versus-host disease. Lactobacillus probiotics as single species or combination probiotic products have been tested for preventing Clostridium difficile–induced gastroenteritis, a common nosocomial and community-based medical condition that has had increased incidence, morbidity, and mortality in past years. Treatment with antibiotics results in disturbance of the GI flora, which is associated with diarrhea and abdominal discomfort in a variable fraction of patients, depending on the age group and the antibiotic used; approximately 15–30% of patients experience recurrence of symptoms after discontinuation of antibiotics. A treatment period with antibiotics only temporarily changes the composition of the microbiota, causing environmental changes. In most cases the cause of the diarrhea is unknown, but a varying proportion of the cases are caused by Clostridium difficile. The C. difficile toxins may cause anything from mild diarrhea, which can be cured simply by terminating the antibiotic treatment, to the life-threatening disease pseudomembranous colitis [20]. Probiotics may also maintain or restore gut microecology during or after antibiotic treatment; Bifidobacteria or Lactobacilli will decrease the duration and/or severity of acute diarrheal disease in infants and children [21].
2.5. Toxicities:
Studies in humans
A number of short-term clinical trials on healthy volunteers attest to the safety of current probiotics. In most studies it is only mentioned that the probiotics did not induce more adverse effects than the placebo or that their tolerance was excellent. In some studies, the presence or absence of GIT disorders has been especially studied, which seems rational because the first and probably only contact between bio-products and the host occur in the GIT [22]. In a few studies, biological parameters were analyzed because it was thought that the probiotics might have an influence on them.
References:
1. Guay, D. R. P. (2009). Cranberry and urinary tract infections. Drugs, 69(7), 775–807.https://doi.org/10.2165/00003495-200969070-00002
2. Kusters, J.G., van Vliet, A.H., Kuipers, E.J., 2006. Pathogenesis of Helicobacter pylori infection. Clin. Microbiol. Rev. 19 (3), 449–490.
3. Burger, O., Ofek, I., Tabak, M., et al., 2000. A high molecular mass constituent of cranberry juice inhibits helicobacter pylori adhesion to human gastric mucus. FEMS Immunol. Med. Microbiol. 29 (4), 295–301.
4. Wittschier, N., Faller, G., Hensel, A., 2007. An extract of Pelargonium sidoides (Eps 7630) inhibits in situ adhesion of Helicobacter pylori to human stomach. Phytomedicine: Int. J. Phytother. Phytopharmacol. 14 (4), 285–288.
5. Wittschier, N., Faller, G., Hensel, A., 2009. Aqueous extracts and polysaccharides from liquorice roots (Glycyrrhiza glabra L.) inhibit adhesion of Helicobacter pylori to human gastric mucosa. J. Ethnopharmacol. 125 (2), 218–223.
6. Douglas, L., 1911. The Bacillus of Long Life. G.P. Putnam’s Sons,New York, US.
7. Fuller, R., 1992. History and development of probiotics. In: Fuller, R. (Ed.), Probiotics. The Scientific Basis Chapman and Hall, London, pp. 1–8.
8. Havenaar, R., Ten Brink, B., Huis in’t Veld, J.H.J., 1992. Selection of strains for probiotic use. In: Fuller, R. (Ed.), Probiotics. The Scientific Basis Chapman and Hall, London, pp. 209–224.
9. FAO/WHO, 2002. Joint FAO/WHO Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food, London Ontario, Canada, April 30 and May 1, 2002, pp. 1–11.
10. Sullivan, A., Nord, C.E., 2002. Probiotics in human infections. J. Antimicrob. Chemother 50, 625–627.
11. Hamilton-Miller, J.M.T., 2003. The role of probiotics in the treatment and prevention of Helicobacter pylori infection. Int. J. Antimicrob. Agents 22, 360–366.
12. Saier, M.H., Mansour, N.M., 2005. Probiotics and prebiotics in human. J. Mol. Microbiol. Biotechnol. 10, 22–25.
13. Anadón, A., Martínez-Larrañaga, M.R., Arés, I., et al., 2016. Prebiotics and probiotics: an assessment of their safety and health benefits. In: Watson, R.R., Preedy, V.R. (Eds.), Bioactive Foods in Promoting Health: Probiotics and Prebiotics. Elsevier Inc, Academic Press, Oxford, UK, pp. 1–23.
14. Mercenier, A., Pavan, S., Pot, B., 2003. Probiotics as biotherapeutic agents: present knowledge and future prospects. Curr. Pharm. Des. 9, 175–191.
15. Tomasik, P.J., Tomasik, P., 2003. Probiotics and prebiotics. Cereal Chem. 80, 113–117.
16. Mercenier, A., Pavan, S., Pot, B., 2003. Probiotics as biotherapeutic agents: present knowledge and future prospects. Curr. Pharm. Des. 9, 175–191.
17. Ciorba, M.A., 2012. A gastroenterologist’s guide to probiotics. Clin. Gastroenterol. Hepatol. 10, 960–968.
18. Mäyrä-MäKinen, A., Bigret, M., 1993. Industrial use and production of lactic acid bacteria. In: Salminen, S., von Wright, A. (Eds.), Lactic Acid Bacteria Marcel Dekker, Inc., New York, NY, pp. 65–96.
19. Goldin, B.R., Gorbach, S.L., 2008. Clinical indications for probiotics: an overview. Clin. Infect. Dis. 46 (Suppl. 2), S96–S100.
20. Vanderhoof, J.A., Whitney, D.B., Antonson, D.L., et al., 1999. Lactobacillus GG in the prevention of antibiotic-associated diarrhea in children. J. Pediat. 135, 564–568.
21. Oberhelman, R.A., Gilman, R.H., Sheen, P., et al., 1999. A placebocontrolled trial of Lactobacillus GG to prevent diarrhea in undernourished Peruvian children. J. Paediatrics 134, 15–20.
22. Salminen, S., von Wright, A., 1998. Current probiotic-safety assured? Microb. Ecol. Health Dis. 10, 68–77.
Valmar
Cap Valmar
(Melatonin, 5-HTP, Pyridoxine & more)
Brand: Valmar
Generic: Melatonin, 5-HTP, Pyridoxine & more
Class: Medical Food Supplement
Route of administration: Oral
Dosage form: Capsule / Syrup
Dose: Capsule : Syrup
Contraindications: Pregnancy: Cat. C
Pack Size: 30 caps.
Price: Capsule: Rs. 410/- | Syrup: Rs. 170/-
Supplement facts:
Supplement Amount per serving
Melatonin 3mg
5-HTP 50mg
Pyridoxine 2mg
Valmar Blend (Valerian,GABA,L-Glycine,Passion flower, Lemon balm, L-theanine, English lavender) 519mg
1. Melatonin
Melatonin is an indoleamine with the chemical name N-acetyl-5-methoxytryptamine and is found in animals, plants, and bacteria. It has endocrine and antioxidant properties in both animals and plants. It is biosynthesized from the neurotransmitter serotonin, which in turn is made from the essential amino acid tryptophan. Because its normally charged amino group is acetylated and a methoxy rather than a hydroxyl group is present in the 5 position, melatonin is a relatively nonpolar compound that readily crosses the blood–brain barrier (BBB) and easily enters cells of all types. In mammals, melatonin has several roles; it is involved in circadian regulation and modulation of reproductive and immune responses, mood, and aging. It is produced by the pineal gland and several peripheral tissues, notably the gut [1]. Because it is liberated into the bloodstream from the pineal and acts through receptors located in a wide variety of body tissues, it can be considered a systemic hormone. Levels of melatonin vary diurnally, seasonally in some mammals, and throughout the life cycle. They are highest during hours of darkness and are lowest among the elderly. This chapter focuses on two areas: (i) recent findings suggesting a variety of therapeutic applications—ranging from improvement of disturbed sleep patterns and mood disorders to adjuvant cancer treatment and surgery and fertility enhancement and (ii) support for the safety of exogenously supplied melatonin. The potential molecular mechanisms underlying these potentially clinically beneficial uses are also discussed.
1.2. Range Of Conditions Where Melatonin May Have Clinical Utility
The evidence for the beneficial properties of melatonin in the derangements described is often derived from studies on animal models of disease, but any relevant human findings are also added to each section.
1.2.1. Disturbances of Sleep Patterns
Melatonin is perhaps most well-known for its ability to induce sleep. Plasma levels are highest during hours of darkness and decline rapidly at the onset of light. Thus, melatonin is widely used to reset normal circadian sleep patterns after disruption by jet lag. Sleep quality can be defined by tiredness upon waking, feeling rested, and the number of awakenings experienced during the night. This can be partly correlated with objective polygraphic measures to determine the extent of fragmentation of sleep. Melatonin has been found to improve sleep quality in patients with a variety of diseases. A dosage of 3 mg/day taken at bedtime for 24 days in a group of 2,062 patients with chronic cerebral ischemia improved multiple types of subjective sleep characteristics [2]. An analysis was made combining results from four clinical trials studying the effects in 401 hypertensive patients aged 55 years or older. The analysis determined that 2 mg/ day of prolonged-release melatonin taken 2 h before bedtime for 3 weeks improved quality of sleep and morning alertness [3]. The improvements in sleep quality were maintained during a follow-up period of 6 months; moreover, the rate of adverse events was lower in the patients using melatonin. Although melatonin had no significant effect on blood pressure in this study, a meta-analysis of seven other clinical trials demonstrated that 2–3 mg/day of controlled-release (but not fast-release) melatonin over periods of 28–90 days significantly reduced nocturnal blood pressure in a variety of subjects, including those with hypertension [4]. In a study of 36 type 2 diabetic individuals, a regimen of 2 mg/day of prolonged-release melatonin taken 2 h before bedtime for 3 weeks resulted in significant improvement in sleep efficiency, wake time after sleep onset, and reduced number of awakenings [5].
The melatonin receptor agonist ramelteon has been used to treat chronic insomnia clinically with success [6]. Some studies demonstrate that melatonin improves sleep quality more modestly. In a recent review of 35 randomized clinical trials, use of melatonin by healthy adults was found to show limited promise for preventing phase shifts from jet lag and for improving insomnia; both healthy adults and insomniacs benefited. However, available data could not confirm a positive benefit for either the initiation of sleep or sleep efficacy [7]. Aging is associated with an altered sleep profile; sleep time is shortened and sleep is more disrupted. Sleeplessness is often found with cardiovascular disease, obesity, type 2 diabetes, cancer, and a range of inflammatory disease states. There is evidence that melatonin can be of use in amelioration of the disrupted sleep associated with Alzheimer’s disease (AD) [8], but this is disputed [9].
Melatonin has also found utility in hypertensive patients in the treatment of sleep disruption caused by depression of intrinsic melatonin production by beta-blockers [10]. Reports of the add-on utility of melatonin to the treatment of several disorders, including epilepsy [11], may primarily be attributable to the improved quality of the sleep–wake cycle [12], but melatonin has also been shown to directly potentiate the anticonvulsant efficacy of phenobarbital [13]. Melatonin is effective as an adjuvant for improving cognitive function and sleep quality impaired by a number of disorders. A study group of 73 outpatients with mild to moderate AD took 2 mg prolonged-release melatonin or placebo daily 1–2 h before bedtime for with myofascial temporomandibular disorder (TMD) and pain [14].
1.2.2. Melatonin and Mood Disorders
The clinical applicability of melatonin and pharmacological agents active at its receptors to depressive and anxiety mood disorders has been reported. The principal antidepressant drug used is agomelatine, which acts as a melatonin MT1 and MT2 receptor agonist. However, agomelatine also acts as a serotonergic 5-HT(2C) receptor antagonist, and this could also contribute its antidepressant activity [15]. Ramelteon, another MT1/MT2 agonist, has no serotonergic antagonistic activity but retains antidepressant properties [16].
The mechanism of action may involve both restoration of a normal sleep cycle and the intrinsic anxiolytic and analgesic properties of melatonin, which has opioid activity [17]. Ramelteon has also been reported to protect against delirium [18]. The biochemical and behavioral deficits found in animal models of several neurodegenerative diseases, including AD, Parkinson’s disease (PD), and cerebral stroke, are mitigated by melatonin treatment [19]. However, reports involving melatonin intervention in these diseases in human trials are very limited. Restoration of deranged sleep patterns may account for the majority of benefits seen in clinical testing of melatonin [20].
Melatonin has shown promise for managing some troublesome side effects of drugs used to treat mood disorders. In a study of schizophrenic patients, treatment with 3 mg/day of melatonin for 8 weeks reduced the metabolic side effects of weight gain, abdominal obesity, and hypertriglyceridemia induced by olanzapine [21]. However, 20 mg/day of controlled-release melatonin for 1 month was no more effective than placebo for reducing dependence on benzodiazepines in a group of 92 elderly outpatients [22].
1.2.3. Slowing Of Common Age-Related Processes
Aging is associated with an increasingly elevated level of inflammatory events [23]. Inappropriate and excessive immune responses characterize many diseases associated with aging, including a range of cardiovascular and neurodegenerative disorders. This heightened level of inflammation appears to be unprovoked by exogenous agents andmay reflect the inappropriate continuation of earlier and more relevant immune responses [24]. In the nervous system, many of the genes whose expression is elevated with age relate to immune function, and melatonin treatment has been shown in aged experimental animals to reverse this trend and restore a more youthful pattern of mRNA production [25]. This is also reflected by reversal of age-associated morphological and biochemical changes in brain [26]. Another important feature of brain aging where melatonin may be of value concerns adult neurogenesis, which has significance that is increasingly being acknowledged. Diminished neurogenesis precedes old age [27], and this decline can be delayed by supplementation with melatonin [28]. In addition, the maintenance of dendritic complexity is enhanced by melatonin [29]. Studies on melatonin in aging humans are relatively scarce, but several promising reports exist. Daily use of 3 mg melatonin protected the retina by delaying macular degeneration—a leading cause of severe visual loss in older people [30]. Even low doses of melatonin used during the evening increased daytime activity of healthy elderly people [31].
1.2.4. Melatonin and Disease Related To Immune Function
Melatonin has also been found to be of utility in the treatment of other diseases and adverse health states in which excess inflammation may not constitute a major element of pathogenesis but where immune function is likely a factor, including cancer [32]. Supplementation of the diet of aged mice with melatonin leads to a major reduction of tumor incidence [33]. Although melatonin is generally reported as supporting cell survival, it appears to promote apoptosis in malignant cells [34]. Several meta-analyses have shown that adjuvant treatment of cancer—particularly solid tumor cancers—with melatonin significantly improves outcomes [35]. Similarly, two recent case studies have reported encouraging results in patients with breast cancer [36] and hepatocellular carcinoma [37]. Encouraging but modest benefits were reported very recently in a clinical trial involving 151 advanced nonsmall cell lung cancer patients also receiving chemotherapy. In this study, 10 or 20 mg/day of melatonin taken at night for 6 months following initiation of chemotherapy decreased DNA damage and tended to improve quality of life; however, even though the longest-living survivors were among those patients receiving melatonin, supplementation did not lengthen survival time significantly overall [38]. Melatonin therapy can also speed the rate of healing of diverse types of wound [39]. In an experimental model of multiple sclerosis—which certainly reflects inappropriate immune responses—melatonin was able to reverse demyelination [40].
1.2.5. Melatonin and Oxidative Stress
There are several states that are predominantly characterized by excessive generation of reactive oxidant species. These include strenuous exercise and chronic pulmonary obstructive disease. Melatonin has been shown to be able to reduce indices of free radical damage in each of these in humans [41].
The relatively low tissue content of active unconjugated melatonin is approximately 1 pM [42]
, which makes a direct antioxidant effect unlikely because several other potent antioxidants such as water-soluble glutathione and lipophilic α-tocopherol are present at much higher intracellular concentrations. Nonetheless, melatonin has been shown to reduce indices of oxidative stress in several clinical situations, including metabolic syndrome [43], Duchenne muscular dystrophy [44], and in severely ill children [45].
1.2.6. Melatonin and Nociception
Melatonin may be effective for reducing chronic pain associated with some diseases. Being antinociceptive, it can act to support conventional anesthetics [46]. In a randomized, placebo-controlled clinical trial involving 32 patients with myofascial TMD pain, pain levels evaluated by two different measurements were reduced in those using 5 mg/day at bedtime over a course of 4 weeks; moreover, these patients also reported requiring smaller and smaller amounts of other analgesics to cope with their pain as the trial progressed [47]..
1.2.7. Reduced toxicity of venoms and pharmacological agents after Melatonin administration
Melatonin has been found to ameliorate the venominduced hemorrhage and myonecrosis incurred after a snake bite [48].
. It is also protective against the nephrotoxicity of tenofovir, a reversetranscriptase inhibitor used in the treatment of HIV infection [49]. The toxicity of several antineoplastics, such as neocarbazine and cyclophosphamide, is reduced in the presence of melatonin [50].
1.2.8. Beneficial Effects of Melatonin Use During Surgical Procedures
Melatonin improves survival of animals after heart transplantation both in the presence and absence of cyclosporine [51]. Prior administration of melatonin also improved survival rates after kidney transplantation [52].
. The persistence of ovaries following transplantation was also increased [53]. Clinical studies in these areas are very limited, but melatonin had only marginally beneficial effects in major liver resection surgery [54]. However, it has been shown to be nearly as effective as clonidine as an agent for sleep induction preceding surgical anesthesia in children aged 1–5 years [55]. Moreover, melatonin administration has been reported to lead to improved outcome following organ transplant procedures [56] and neonatal surgery [57]. The more successful reports used much higher doses of melatonin. Melatonin appears to have clinical utility in reducing the damage incurred by ischemia-reperfusion injury in the liver, which is very susceptible to such fluxes in vascular supply [58]. However, several trials on the potential of melatonin to mitigate the effects of reperfusion injury on the heart have not been proven to be successful [59].
1.2.9. Migraine Amelioration by Melatonin
Melatonin shows promise for reducing severity of migraine headaches in adults and children. In a recent study of 60 children (mean age, 10.3 years), monthly frequency of migraines was reduced by 55%, duration was reduced by 51%, and severity was reduced by 43%; the most frequent side effect was daytime sleepiness that occurred in seven subjects [60].
1.2.10. Melatonin Supplementation and Fertility
The quality of oocytes used for in vitro fertilization has been reported to be improved by treatment of the donors with melatonin [61]. The efficacy of oral melatonin supplementation on oocyte and embryo quality in patients in an assisted reproductive technologies program has been studied [62]. Patients were treated with 3 mg/day melatonin for at least 2 weeks. To evaluate the cumulative effect of melatonin supplementation, cycle outcomes between the first (no supplementation) and second cycles (melatonin supplementation) of patients who completed two treatment cycles were compared.
There were no significant differences in maturation rates, blastocyst rates, and the rate of good-quality blastocysts between the first and second cycles. However, melatonin increased the fertilization rate from 35.1% to 68.2% and the proportion of good-quality embryos from 48.0% to 65.6%; these effects were ascribed to a reduction in oxidative damage.
1.3. Safety of Melatonin
The term nutraceutical implies that a substance has little or no toxicity even when consumed for long periods of time. Although melatonin is considered to have little toxicity, much of the evidence for this comes from short-term studies [63]. In attempting to assess its safety as a nutraceutical, in addition to considering short-term toxicity data, a careful evaluation of any information pertaining to melatonin’s long-term consumption should be performed. The only adverse report of potential harmfulness of melatonin is a report indicating that, in an isolated system, its metabolite 6-hydroxymelatonin promoted metalion induced lesions to guanine and thymidine residues in DNA [64]. Most redox-capable antioxidants such as ascorbic acid and lipoic acid can also facilitate this cycling. Neither short-term treatment nor extended treatment of humans with melatonin has led to symptoms of dependence, tolerance, rebound insomnia, or withdrawal [65]. With respect to evaluating the safety of melatonin, it is important to distinguish two possible applications: its low-dose nutraceutical consumption by relatively healthy individuals and the acute administration of high dosages to patients with serious medical conditions. The former involves the oral, possibly chronic, intake of relatively low dosages (0.3–10 mg/day), anticipated to be taken at bedtime or when the induction of nocturnal physiological conditions is desired. Frequently, the goal is the restoration or modulation of normal homeostatic function to compensate for age-related changes or circadian cycle disruption, rather than as treatment of serious disease. Here, safety considerations need to focus on the presence or lack of association of low-dose, possibly chronic, consumption of melatonin with increased incidence of disease or pathology in humans. Although high-quality, long-term clinical studies would provide the best evidence for or against such an association, the lack of such studies means that one has to rely on shorterterm, less powerful ones. Nevertheless, the safety and toxicity of melatonin have been the focus of a number of studies, and this hormone is frequently characterized as being both safe and nontoxic [66]. The safety of melatonin used as a nutraceutical ingredient in food has been challenged on at least one occasion in a Food and Drug Administration (FDA) warning letter [67]. Rather than restate evidence for melatonin’s safety, insight from a different perspective may be gained by scrutinizing—at least as an illustrative case—the opposing evidence provided in this document in which it may be presumed that the strongest available references would be selected for characterizing melatonin as being toxic and/or unsafe. The warning letter cited 23 reports that “have raised safety concerns” about melatonin. Of these, the entire text of 21 could be accessed. Surprisingly, terms such as “safe,” “safety,” “toxic,” “toxicity,” or “adverse” did not occur at all in these 21 or they occurred in statements actually supporting the safety and/or nontoxicity of melatonin. Most of these references provide poor or no support for safety concerns either in humans or in the experimental models studied in rodents or in vitro. Among the references cited as raising concerns over melatonin’s effects on blood glucose homeostasis, two of the studies [68] performed no glucose measurements and mentioned no concerns; a third study in rats [69] found that melatonin induced no change in blood glucose. A fourth study [70] reported decreased glucose tolerance and insulin sensitivity in older women treated with 1 mg of melatonin in the morning. Typically, longterm supplementation of melatonin is recommended to be used only at bedtime, and not in the morning. As noted by the study’s authors, the effects measured may merely represent the induction during the daytime of the reduced nocturnal glucose sensitivity observed in normal, healthy humans. In contrast to the results of this study, beneficial improvements in HbA1c and no such possibly deleterious effects on glucose metabolism were found in a more recent study of diabetic patients [71]. There were two adverse reports among studies cited to support reproductive concerns [72] observed strain-specific mortality in rat pups when dams received a 250-times higher dose (on a weight basis, for a 75-kg human) than the 3 mg/day frequently recommended for humans. The strain-specificity and high dosage suggest these results may not be relevant to typical human nutraceutical usage. A second study reporting reduced semen quality in men receiving 3 mg/day of melatonin for 3 months [73] is of more concern. However, this reduction occurred in only two of eight subjects, suggesting that only a minority of males may be affected; in any case, a much larger study is required for confirmation. With regard to females, an in vitro study merely exposed cultured human granulosa cells to melatonin without expressing any safety implications [74]. Additional human studies in which no safety concerns were mentioned included those of [75] and [76], but only the abstracts could be checked. Rodents were used in seven of the reports cited in the FDA warning letter. In rodents, adverse effects were observed only at melatonin concentrations much higher than the 0.5–10 mg typically suggested for long-term nutraceutical consumption in humans. Moreover, all but one of the rodent experiments were performed on animals that modeled human disease, and thus they may have questionable applicability to healthy humans. For example, [77] studied castrated rats without adverse effects, whereas [78] reported that under normal conditions, increased retinal cell death and thinned outer nuclear layer in retinas of non-pigmented rats were associated with administration of 100 times the melatonin dosage typical of human intake. Because melatonin may have greatly different effects in humans compared to rodents in certain circumstances [79] , caution needs to be used in applying the results of animal studies when evaluating the safety of melatonin consumption by humans [80] showed that large amounts of melatonin fed to mice in an atherogenic diet exacerbate plaque formation, but they state that “melatonin had no toxicity on animal health even at this high dose” and express caution by noting that one cannot infer “that high melatonin doses (in mice) would have any deleterious effect on atherosclerosis development in humans.” In humans, other vascular studies—those of [81],—expressed no safety concerns. Similarly, authors of a vision-related study in humans [82] expressed no concerns about melatonin’s safety. Safety concerns of melatonin may apply when treating patients with specific medical conditions, but not in healthy humans using melatonin at bedtime. [83] .reported that a single 15 mg dose of melatonin administered during the daytime lowered input to subjects’ retinal cones; no safety concerns were raised other than cautioning that melatonin should not be taken during the daytime, and the authors concluded that this effect “may serve to promote (normal) night vision.” Further suggesting that undesirable effects may not be a concern in healthy individuals, serious adverse events were reported only in studies of small numbers of human subjects with medical conditions. In this regard, [84] reported that exogenous melatonin, in conjunction with late-night bright light, exacerbated symptoms in eight patients with restless legs syndrome, and [85] observed an increased incidence of seizures among six children with neurological disabilities who had undergone multiple medical procedures. In summary, this examination of references supporting the “safety concerns” raised against melatonin supports the notion that there is little evidence engendering significant generalized concerns about melatonin’s safety, particularly with regard to typically recommended dosages of melatonin consumption in healthy humans. There is no doubt that the toxicity of melatonin is extremely low. Thus, even 800 mg/kg body weight has not been proven lethal in experimental animals [86]. Few nutraceutical vitamins or cofactors have such a large margin of safety.
2. l-Theanine
Tea is often used as a relaxing beverage. l-Theanine is an amino acid extracted from green or black tea. In the brain, l-theanine increases dopamine, serotonin, and the inhibitory neurotransmitter glycine [87], [88] have reported inhibitory effects of higher doses of theanine on caffeine stimulation evaluated by electroencephalogram (EEG) in the rat. However, when given by itself, lower doses of theanine resulted in excitatory effects, suggesting a dual activity of theanine depending on the dose [89] have reported that even lower doses of l-theanine can induce alpha-wave production as observed in EEG tracings from 54 healthy participants at baseline and at 45, 60, 75, 90, and 105 min after 50 mg l-theanine or placebo. Statistical analysis of data showed a significantly greater increase in alpha-wave production in the theanine group than in the placebo group [90] have reported that the acute stress response elicited by a math test was attenuated by 200 mg theanine, as assessed by heart rate and salivary IgA.
3. Gamma Aminobutyric Acid
GABA is an inhibitory neurotransmitter widely distributed throughout the CNS. Too much excitation can lead to irritability, restlessness, insomnia, seizures, and movement disorders, so it must be balanced with inhibition that is provided by GABA acting like a “brake” during times of stress. Medications for anxiety, such as benzodiazepines, stimulate GABA receptors and induce relaxation. Either low GABA levels or decreased GABA function in the brain is associated with several psychiatric and neurological disorders, including anxiety, depression, insomnia, and epilepsy. Studies indicate GABA can improve relaxation and enhance sleep. Because of the association between low GABA levels and these conditions, many antianxiety and sleepenhancing drugs have been developed that interact primarily with GABA receptors. These include the benzodiazepine drugs—alprazolam, diazepam, flurazepam, quazepam, temazepam, and triazolam—and zolpidem tartrate and baclofen. GABA mediates presynaptic inhibition of primary afferent fibers in the motor system. It regulates brain excitability via GABAA receptors, which are classified into three major groups (alpha, beta, and gamma) with subunits that determine its pharmacological activity. GABAA receptors are highly expressed in the thalamus, a region of the brain involved with sleep processes [91] have reported that gabapentin (structurally similar to GABA; increases brain GABA levels) has been found to be effective for panic disorder and improves sleep disturbances associated with alcohol consumption [92] showed GABA-agonist drugs are sedatives used to treat insomnia. Natural therapies that produce relaxation also act, at least in part, by enhancing GABA levels [93] have reported that valerenic acid potentiates and inhibits GABAA receptors [94] have reported reduced GABAA receptor binding in veterans with posttraumatic stress disorder as demonstrated by positron emission tomography scan. On EEG, alpha waves are generated in a relaxed state, whereas beta waves are seen in stressful situations that make mental concentration difficult. Therefore, the ratio of alpha to beta waves is used as an indication of relaxation and better concentration. In general, the greater the alphato- beta ratio, the more relaxed and alert the person is. [95] have reported relaxation and immunity enhancement effects of GABA administration in humans, and it was seen that GABA produced significant effects on increasing alpha waves and on decreasing beta waves, resulting in a significant increase in the alpha-to-beta wave ratio. Due to its relaxation effects, GABA may be considered to be a sleep aid.
4. l-Tryptophan/5-Hydroxytryptophan (5-HTP)
l-Tryptophan, a large neutral amino acid essential to human metabolism, is the metabolic precursor of serotonin (a neurotransmitter), melatonin (a neurohormone), and niacin (vitamin B3). Tryptophan hydroxylase is the rate-limiting enzyme for serotonin production and involves the conversion of tryptophan to 5-HTP. This enzyme can be inhibited by stress, insulin resistance, magnesium or vitamin B6 deficiency, or increasing age [96] have demonstrated the inhibitory effect of 5-HTP on cholecystokinin-4-induced panic attacks in healthy volunteers. Another study conducted by [97] examined that acute l-5-HTP administration inhibits panic attacks induced by carbon dioxide in panic disorder patients, as displayed by reduced panic symptom scores and number of panic attacks in comparison to placebo [98] have reported that l-5-HT treatment helps in reduction of sleep terrors in children because of its enhancement of serotonin and melatonin as compared to the untreated group.
5. Pyridoxal 5′-Phosphate (Active Vitamin B6)
Pyridoxal 5-phosphate (the active form of vitamin B6) is a necessary cofactor for the formation of several important neurotransmitters associated with stress. Within the brain, glutamic acid is converted to GABA via the enzyme glutamate decarboxylase and its cofactor pyridoxal 5′-phosphate. GABA is metabolized by gamma aminobutyrate transaminase, also a P5Pdependent enzyme, forming an intermediate metabolite succinate semialdehyde. P5P is a cofactor in the conversion of 5-HTP to serotonin. Furthermore, conversion of l-tryptophan to 5-HTP, the rate-limiting step in serotonin synthesis, can be inhibited by stress, insulin resistance, magnesium or vitamin B6 deficiency, or increasing age. The decarboxylation of 5-HTP to serotonin is dependent on the presence of pyridoxal 5′-phosphate. P5P is also a cofactor in the synthesis of dopa to dopamine in the pathway converting tyrosine to epinephrine and norepinephrine [99].
References:
1. Chen, C.Q., Fichna, J., Bashashati, M., et al., 2011. Distribution, function and physiological role of melatonin in the lower gut. World J. Gastroenterol. 17, 3888–3898
2. Poluéktov, M.G., Levin, I.I., Boĭko, A.N., et al., 2012. The results of Russian multicenter open-label observational study of the effic_ cacy and safety of мelaxen (melatonin) for the treatment of disordered sleep in patients with chronic cerebral ischemia. Zh. Nevrol. Psikhiatr. Im. S S Korsakova 112, 26–31
3. Lemoine, P., Wade, A., Katz, A., et al., 2012. Efficacy and safety of prolonged-release melatonin for insomnia in middle-aged and elderly patients with hypertension: a combined analysis of controlled clinical trials. Integr. Blood Press Control. 5, 9–17
4. Grossman, E., Laudon, M., Zisapel, N., 2011. Effect of melatonin on nocturnal blood pressure: meta-analysis of randomized controlled trials. Vasc. Health Risk Manag. 7, 577–584
5. Garfinkel, D., Zorin, M., Wainstein, J., et al., 2011. Efficacy and safety of prolonged-release melatonin in insomnia patients with diabetes: a randomized, double-blind, crossover study. Diabetes Metab. Syndr. Obes. 4, 307–313
6. Liu, J., Wang, L.N., 2012. Ramelteon in the treatment of chronic insomnia: systematic review and meta-analysis. Int. J. Clin. Pract. 66, 867–873
7. Costello, R.B., Lentino, C.V., Boyd, C.C., et al., 2014. The effectiveness of melatonin for promoting healthy sleep: a rapid evidence assessment of the literature. Nutr. J. 13, 106
8. Cardinali, D.P., Furio, A.M., Brusco, L., 2011. The use of chronobiotics in the resynchronization of the sleep/wake cycle. Therapeutical application in the early phases of Alzheimer’s disease. Recent Pat. Endocr. Metab. Immune Drug Discov. 5, 80–90
9. McCleery, J., Cohen, D.A., Sharpley, A.L., 2014. Pharmacotherapies for sleep disturbances in Alzheimer’s disease. Cochrane Database Syst. Rev. 3, CD009178
10. Scheer, F.A., Morris, C.J., Garcia, J.I., et al., 2012. Repeated melatonin supplementation improves sleep in hypertensive patients treated with beta-blockers: a randomized controlled trial. Sleep 35, 1395–1402
11. Goldberg-Stern, H., Oren, H., Peled, N., et al., 2012. Effect of melatonin on seizure frequency in intractable epilepsy: a pilot study. J. Child Neurol. 27, 1524–1528
12. Gupta, M., Aneja, S., Kohli, K., 2004. Add-on melatonin improves quality of life in epileptic children on valproate monotherapy: a randomized, double-blind, placebo-controlled trial. Epilepsy Behav. 5, 316–321
13. Forcelli, P.A., Soper, C., Duckles, A., et al., 2013. Melatonin potentiates the anticonvulsant action of phenobarbital in neonatal rats. Epilepsy Res. 107, 217–223
14. Vidor, L., Torres, I., Custódio de Souza, I., et al., 2013. Analgesic and sedative effects of melatonin in temporomandibular disorders: a double-blind, randomized, parallel-group, placebo-controlled study. J. Pain Symptom Manage. 46, 422–432
15. Fuchs, E., Simon, M., Schmelting, B., 2006. Pharmacology of a new antidepressant: benefit of the implication of the melatonergic system. Int. Clin. Psychopharmacol. 21 (Suppl. 1), S17–S20
16. Hardeland, R., Poeggeler, B., 2012. Melatonin and synthetic melatonergic agonists: actions and metabolism in the central nervous system. Cent. Nerv. Syst. Agents Med. Chem. 12, 189–216
17. Srinivasan, V., Pandi-Perumal, S.R., Spence, D.W., et al., 2010. Potential use of melatonergic drugs in analgesia: mechanisms of action. Brain Res. Bull. 81, 362–371
18. Hatta, K., Kishi, Y., Wada, K., et al., 2014. Preventive effects of ramelteon on delirium: a randomized placebo-controlled trial. JAMA Psychiatry 71, 397–403
19. Patki, G., Lau, Y.S., 2011. Melatonin protects against neurobehavioral and mitochondrial deficits in a chronic mouse model of Parkinson’s disease. Pharmacol. Biochem. Behav. 99, 704–711.
20. Rothman, S.M., Mattson, M.P., 2012. Sleep disturbances in Alzheimer’s and Parkinson’s diseases. Neuromolecular Med. 14, 194–204.
21. Modabbernia, A., Heidari, P., Soleimani, R., et al., 2014. Melatonin for prevention of metabolic side-effects of olanzapine in patients with first-episode schizophrenia: randomized double-blind placebo controlled study. J. Psychiatr. Res. 53, 133–140
22. Lähteenmäki, R., Puustinen, J., Vahlberg, T., et al., 2014. Melatonin for sedative withdrawal in older patients with primary insomnia: a randomized double-blind placebo-controlled trial. Br. J. Clin. Pharmacol. 77, 975–985
23. Bondy, S.C., Sharman, E.H., 2010. Melatonin, oxidative stress and the aging brain. In: Bondy, S.C., Maiese, K. (Eds.), Oxidative Stress in Basic Research and Clinical Practice: Aging and Age-Related Disorders Humana Press, Totowa, NJ, pp. 339–357
24. Qin, L., Wu, X., Block, M.L., et al., 2007. Systemic LPS causes chronic neuro inflammation and progressive neurodegeneration. Glia 55, 453–462
25. Sharman, E.H., Bondy, S.C., Sharman, K.Z., et al., 2007. Effects of melatonin and age on gene expression in mouse CNS using microarray analysis. Neurochem. Int. 50, 336–344
26. Zhou, J., Yang, F., Zhou, L., et al., 2014. Dietary melatonin attenuates age-related changes in morphology and in levels of key proteins in globus pallidus of mouse brain. Curr. Aging Sci. 5, 242–253
27. Leuner, B., Kozorovitskiy, Y., Gross, C.G., et al., 2007. Diminished adult neurogenesis in the marmoset brain precedes old age. Proc. Natl. Acad. Sci. U.S.A. 104, 17169–17173
28. Ramirez-Rodriguez, G., Vega-Rivera, N.M., Benitez-King, G., et al., 2012. Melatonin supplementation delays the decline of adult hippocampal neurogenesis during normal aging of mice. Neurosci. Lett. 530, 53–58
29. Ramirez-Rodriguez, G., Ortiz-Lopez, L., Dominguez-Alonso, A., et al., 2011. Chronic treatment with melatonin stimulates dendrite maturation and complexity in adult hippocampal neurogenesis of mice. J. Pineal Res. 50, 29–37
30. Yi, C., Pan, X., Yan, H., et al., 2005. Effects of melatonin in age-related macular degeneration. Ann. N.Y. Acad. Sci. 1057, 384–392.
31. Valtonen, M., Niskanen, L., Kangas, A.P., et al., 2005. Effect of melatonin rich night-time milk on sleep and activity in elderly institutionalized subjects. Nord. J. Psychiatry 59, 217–221
32. Cutando, A., López-Valverde, A., Arias-Santiago, S., et al., 2012. Role of melatonin in cancer treatment. Anticancer Res. 32, 2747–2753
33. Sharman, E.H., Sharman, K.Z., Bondy, S.C., 2011. Extended exposure to dietary melatonin reduces tumor number and size in aged mice. Exp. Gerontol. 46, 18–22
34. Sánchez-Hidalgo, M., Guerrero, J.M., Villegas, I., et al., 2012. Melatonin, a natural programmed cell death inducer in cancer. Curr. Med. Chem. 19, 3805–3821
35. Cutando, A., López-Valverde, A., Arias-Santiago, S., et al., 2012. Role of melatonin in cancer treatment. Anticancer Res. 32, 2747–2753
36. Di Bella, G., Mascia, F., Ricchi, A., et al., 2013. Evaluation of the safety and efficacy of the first-line treatment with somatostatin combined with melatonin, retinoids, vitamin D3, and low doses of cyclophosphamide in 20 cases of breast cancer: a preliminary report. Neuro Endocrinol. Lett. 34, 660–668
37. Tomov, B., Popov, D., Tomova, R., et al., 2013. Therapeutic response of untreatable hepatocellular carcinoma after application of the immune modulators IL-2, BCG and melatonin. Anticancer Res. 33, 4531–4535
38. Sookprasert, A., Johns, N., Phunmanee, A., et al., 2014. Melatonin in patients with cancer receiving chemotherapy: a randomized, double- blind, placebo-controlled trial. Anticancer Res. 34, 7327–7337
39. Celinski, K., Konturek, P.C., Konturek, S.J., et al., 2011. Effects of melatonin and tryptophan on healing of gastric and duodenal ulcers with Helicobacter pylori infection in humans. J. Physiol. Pharmacol. 62, 521–526
40. Kashani, I.R., Rajabi, Z., Akbari, M., et al., 2014. Protective effects of melatonin against mitochondrial injury in a mouse model of multiple sclerosis. Exp. Brain Res. 232, 2835–2846
41. Ochoa, J.J., Díaz-Castro, J., Kajarabille, N., et al., 2011. Melatonin supplementation ameliorates oxidative stress and inflammatory signaling induced by strenuous exercise in adult human males. J.Pineal Res. 51, 373–380
42. Lahiri, D.K., Ge, Y.-W., Sharman, E.H., et al., 2004. Age-related changes in serum melatonin in mice, higher levels of combined melatonin and melatonin sulfate in the brain cortex than serum, heart,. liver and kidney tissues. J. Pineal Res. 36, 217–223
43. Koziróg, M., Poliwczak, A.R., Duchnowicz, P., et al., 2011. Melatonin treatment improves blood pressure, lipid profile, and parameters of oxidative stress in patients with metabolic syndrome. J. Pineal Res. 50, 261–266
44. Chahbouni, M., Escames, G., López, L.C., et al., 2011. Melatonin treatment counteracts the hyperoxidative status in erythrocytes of patients suffering from Duchenne muscular dystrophy. Clin. Biochem. 44, 853–858
45. Marseglia, L., Aversa, S., Barberi, I., et al., 2013. High endogenous melatonin levels in critically ill children: a pilot study. J. Pediatr. 162, 357–360
46. Marseglia, L., D’Angelo, G., Manti, S., et al., 2015. Analgesic, anxiolytic and anaesthetic effects of melatonin: new potential uses in pediatrics. Int. J. Mol. Sci. 16, 1209–1220
47. Vidor, L., Torres, I., Custódio de Souza, I., et al., 2013. Analgesic and sedative effects of melatonin in temporomandibular disorders: a double-blind, randomized, parallel-group, placebo-controlled study. J. Pain Symptom Manage. 46, 422–432
48. Katkar, G.D., Sundaram, M.S., Hemshekhar, M., et al., 2014. Melatonin alleviates Echis carinatus venom-induced toxicities by modulating inflammatory mediators and oxidative stress. J. Pineal Res. 56, 295–312
49. Ramamoorthy, H., Abraham, P., Isaac, B., 2014. Preclinical efficacy of melatonin in the amelioration of tenofovir nephrotoxicity by the attenuation of oxidative stress, nitrosative stress, and inflammation in rats. J. Basic Clin. Physiol. Pharmacol. 27, 1–13
50. Alp, B.F., Kesik, V., Malkoç, E., et al., 2014. The effect of melatonin on procarbazine induced testicular toxicity on rats. Syst. Biol. Reprod. Med. 20, 1–6
51. Liu, C., Hong, T., Shao, M., et al., 2014. Melatonin synergized with cyclosporine A improves cardiac allograft survival by suppressing inflammation and apoptosis. Mol. Med. Rep. 10, 1323–1328
52. Li, Z., Nickkholgh, A., Yi, X., et al., 2009. Melatonin protects kidney grafts from ischemia/reperfusion injury through inhibition of NF-kB and apoptosis after experimental kidney transplantation. J. Pineal Res. 46, 365–372
53. Hemadi, M., Shokri, S., Pourmatroud, E., et al., 2012. Follicular dynamic and immunoreactions of the vitrified ovarian graft after host treatment with variable regimens of melatonin. Am. J. Reprod. Immunol. 67, 401–412
54. Nickkholgh, A., Schneider, H., Sobirey, M., et al., 2011. The use of highdose melatonin in liver resection is safe: first clinical experience. J. Pineal Res. 50, 381–388
55. Almenrader, N., Haiberger, R., Passariello, M., 2013. Steal induction in preschool children: is melatonin as good as clonidine? A prospective, randomized study. Paediatr. Anaesth. 23, 328–333
56. Liu, C., Hong, T., Shao, M., et al., 2014. Melatonin synergized with cyclosporine A improves cardiac allograft survival by suppressing inflammation and apoptosis. Mol. Med. Rep. 10, 1323–1328
57. Gitto, E., Romeo, C., Reiter, R.J., et al., 2004. Melatonin reduces oxidative stress in surgical neonates. J. Pediatr. Surg. 39, 184–189.
58. Li, Y., Yang, Y., Feng, Y., et al., 2014. A review of melatonin in hepatic ischemia/reperfusion injury and clinical liver disease. Ann. Med. 46, 503–511
59. Ekeløf, S.V., Halladin, N.L., Jensen, S.E., et al., 2014. Effects of intracoronary melatonin on ischemia-reperfusion injury in ST-elevation myocardial infarction. Heart Vessels [Epub ahead of print] PMID: 25319673
60. Fallah, R., Shoroki, F., Ferdosian, F., 2015. Safety and efficacy of melatonin in pediatric migraine prophylaxis. Curr. Drug Saf. 10 (2), 132–135
61. Rizzo, P., Raffone, E., Benedetto, V., 2010. Effect of the treatment with myo-inositol plus folic acid plus melatonin in comparison with a treatment with myo-inositol plus folic acid on oocyte quality and pregnancy outcome in IVF cycles. A prospective, clinical trial. Eur. Rev. Med. Pharmacol. Sci. 14, 555–561
62. Nishihara, T., Hashimoto, S., Ito, K., et al., 2014. Oral melatonin supplementation improves oocyte and embryo quality in women undergoing in vitro fertilization-embryo transfer. Gynecol. Endocrinol. 30, 359–362
63. Bruni, O., Alonso-Alconada, D., Besag, F., et al., 2015. Current role of melatonin in pediatric neurology: clinical recommendations. Eur. J. Paediatr. Neurol. 19, 122–133
64. Bruni, O., Alonso-Alconada, D., Besag, F., et al., 2015. Current role of melatonin in pediatric neurology: clinical recommendations. Eur. J. Paediatr. Neurol. 19, 122–133
65. Lyseng-Williamson, K.A., 2012. Melatonin prolonged release: in the treatment of insomnia in patients aged ≥55 years. Drugs Aging 29, 911–923
66. Garfinkel, D., Zorin, M., Wainstein, J., et al., 2011. Efficacy and safety of prolonged-release melatonin in insomnia patients with diabetes: a randomized, double-blind, crossover study. Diabetes Metab. Syndr. Obes. 4, 307–313
67. FDA, 2011. Warning Letters: HBB, LLC dba Baked World 7/28/11. Available from: http://www.fda.gov/ICECI/EnforcementActions/ WarningLetters/ucm266129.htm (February 17, 2015)
68. Puchalski, S.S., Green, J.N., Rasmussen, D.D., 2003. Melatonin effect on rat body weight regulation in response to high-fat diet at middle age. Endocrine 21, 163–167
69. Peschke, E., Schucht, H., Mühlbauer, E., 2010. Long-term enteral administration of melatonin reduces plasma insulin and increases expression of pineal insulin receptors in both Wistar and type 2-diabetic Goto-Kakizaki rats. J. Pineal Res. 49, 373–381
70. Cagnacci, A., Arangino, S., Renzi, A., et al., 2001. Influence of melatonin administration on glucose tolerance and insulin sensitivity of postmenopausal women. Clin. Endocrinol. 54, 339–346
71. Garfinkel, D., Zorin, M., Wainstein, J., et al., 2011. Efficacy and safety of prolonged-release melatonin in insomnia patients with diabetes: a randomized, double-blind, crossover study. Diabetes Metab. Syndr. Obes. 4, 307–313
72. Singh, H.J., Keah, L.S., Kumar, A., et al., 2011. Adverse effects of melatonin on rat pups of Wistar-Kyoto dams receiving melatonin supplementation during pregnancy. Exp. Toxicol. Pathol. 64, 751–752
73. Luboshitzky, R., Shen-Orr, Z., Nave, R., et al., 2002. Melatonin administration alters semen quality in healthy men. J. Androl. 23, 572–578
74. Woo, M.M., Tai, C.J., Kang, S.K., et al., 2001. Direct action of melatonin in human granulosa-luteal cells. J. Clin. Endocrinol. Metab. 86, 4789–4797
75. Okatani, Y., Okamoto, K., Hayashi, K., et al., 1998. Maternal-fetal transfer of melatonin in pregnant women near term. J. Pineal Res. 25, 129–134
76. Cagnacci, A., Paoletti, A.M., Soldani, R., et al., 1995a. Melatonin enhances the luteinizing hormone and follicle-stimulating hormone responses to gonadotropin-releasing hormone in the follicular, but not in the luteal, menstrual phase. J. Clin. Endocrinol. Metab. 80, 1095–1099. Cagnacci, A., Soldani, R., Yen, S.S., 1995b. Exogenous melatonin enhances luteinizing hormone levels of women in the follicular but not in the luteal menstrual phase. Fertil. Steril. 63, 996–999
77. Puchalski, S.S., Green, J.N., Rasmussen, D.D., 2003. Melatonin effect on rat body weight regulation in response to high-fat diet at middle age. Endocrine 21, 163–167
78. Wiechmann, A.F., Chignell, C.F., Roberts, J.E., 2007. Influence of dietary melatonin on photoreceptor survival in the rat retina: an ocular toxicity study. Exp. Eye Res. 86, 241–250
79. Peschke, E., Schucht, H., Mühlbauer, E., 2010. Long-term enteral administration of melatonin reduces plasma insulin and increases expression of pineal insulin receptors in both Wistar and type 2-diabetic Goto-Kakizaki rats. J. Pineal Res. 49, 373–381
80. Tailleux, A., Torpier, G., Bonnefont-Rousselot, D., et al., 2002. Daily melatonin supplementation in mice increases atherosclerosis in proximal aorta. Biochem. Biophys. Res. Commun. 293, 1114–1123
81. Cook, J.S., Sauder, C.L., Ray, C.A., 2011. Melatonin differentially affects vascular blood flow in humans. Am. J. Physiol. Heart Circ. Physiol. 300, H670–H674
82. Rufiange, M., Dumont, M., Lachapelle, P., 2002. Correlating retinal function with melatonin secretion in subjects with an early or late circadian phase. Invest. Ophthalmol. Vis. Sci. 43, 2491–2499
83. Gagné, A.M., Danilenko, K.V., Rosolen, S.G., et al., 2009. Impact of oral melatonin on the electroretinogram cone response. J. Circadian Rhythms 7, 14
84. Whittom, S., Dumont, M., Petit, D., et al., 2010. Effects of melatonin and bright light administration on motor and sensory symptoms of RLS. Sleep Med. 11, 351–355
85. Sheldon, S.H., 1998. Pro-convulsant effects of oral melatonin in neurologically disabled children. Lancet 351, 1254
86. Barchas, J., DaCosta, F., Spector, S., 1967. Acute pharmacology of melatonin. Nature 214, 919–920
87. Yamada, T., Terashima, T., Kawano, S., et al., 2009. Theanine, gammaglutamylethylamide, a unique amino acid in tea leaves, modulates neurotransmitter concentrations in the brain striatum interstitium in conscious rats. Amino Acids 36, 21–27
88. Kakuda, T., Nozawa, A., Unno, T., et al., 2000. Inhibiting effects of Theanine on caffeine stimulation evaluated by EEG in the rat. Biosci. Biotechnol. Biochem. 64, 287–293
89. Nobre, A.C., Rao, A., Owen, G.N., 2008. l-Theanine, a natural constituent in tea, and its effect on mental state. Asia Pac. J. Clin. Nutr. 17, 167–168
90. Kimura, K., Ozeki, M., Juneja, L.R., et al., 2007. l-Theanine reduces psychological and physiological stress responses. Biol. Psychol. 74, 39–45
91. Orser, B.A., 2006. Extrasynaptic GABAA receptors are critical targets for sedative-hypnotic drugs. J. Clin. Sleep Med. 2, S12–S18.
92. Roth, T., Soubrane, C., Titeux, L., et al., 2006. Efficacy and safety of zolpidem-MR: a double-blind, placebo-controlled study in adults with primary insomnia. Sleep Med. 7, 397–406
93. Khom, S., Baburin, I., Timin, E., et al., 2007. Valerenic acid potentiates and inhibits GABA(A) receptors: molecular mechanism and subunit specificity. Neuropharmacology 53, 178–187
94. Geuze, E., van Berckel, B.N., Lammertsma, A.A., et al., 2008. Reduced GABAA benzodiazepine receptor binding in veterans with posttraumatic stress disorder. Mol. Psychiatry 13, 74–83
95. Abdou, A.M., Higashiguchi, S., Horie, K., et al., 2006. Relaxation and immunity enhancement effects of gamma aminobutyric acid (GABA) administration in humans. Biofactors 26, 201–208
96. Maron, E., Toru, I., Vasar, V., et al., 2004. The effect of 5-hydroxytryptophan on cholecystokinin-4-induced panic attacks in healthy volunteers. J. Psychopharmacol. 18, 194–199
97. Schruers, K., van Diest, R., Overbeek, T., et al., 2002. Acute l-5- hydroxytryptophan administration inhibits carbon dioxideinduced panic in panic disorder patients. Psychiatry Res. 113, 237–243
98. Bruni, O., Ferri, R., Miano, S., et al., 2004. L-5-Hydroxytryptophan treatment of sleep terrors in children. Eur. J. Pediatr. 163, 402–407
99. van Praag, H.M., Lemus, C., 1986. Monoamine precursors in the treatment of psychiatric disorders. In: Wurtman, R.J., Wurtman, J.J. (Eds.), Nutrition and the Brain Raven Press, New York, NY, pp. 89–139