Echinacea purpurea/angustifolia root | 1:2 | 30 mL |
Andrographis paniculata | 1:2 | 10 mL |
Silybum marianum | 1:1 | 20 mL |
Matricaria recutita | 1:2 | 20 mL |
Filipendula ulmaria | 1:2 | 20 mL |
total | 100 mL |
Dose: 5 mL with water three times a day before meals.
Case history
A female patient aged 35 had been overseas and developed an acute gastrointestinal infection with pain and diarrhoea. The infection passed but on returning home she experienced bloating and diarrhoea often after eating. Stool culture did not demonstrate the presence of an infection. Symptoms could be controlled by restricting her diet to a very simple one: rice and vegetables.
Echinacea angustifolia root | 1:2 | 30 mL |
Picrorrhiza kurroa | 1:2 | 10 mL |
Silybum marianum | 1:1 | 20 mL |
Matricaria chamomilla | 1:2 | 20 mL |
Filipendula ulmaria | 1:2 | 20 mL |
total | 100 mL |
Dose: 5 mL with water three times a day before meals.
Hydrastis tablets 500 mg, one tablet with each meal.
After 4 weeks of treatment, symptoms had improved and she could often be more adventurous with her diet without causing problems. However, symptoms still occurred on many days.
During the next 4 weeks her condition continued to improve with herbal treatment. After another 4 weeks she could eat normally and the herbs were discontinued without adverse effect. Note that Andrographis would be a suitable substitute for the Picrorrhiza in the above prescription.
Constipation is medically defined as a bowel frequency of less than three times a week or the need to strain more than 25% of the time during defaecation. However, it is probably less than optimum for health to defaecate less than once a day. Phytotherapists certainly believe that regular bowel movements are necessary for the maintenance of good health. Constipation may be associated with diseases such as hypothyroidism and Parkinson’s disease and these should always be excluded.
Factors that may cause or contribute to simple constipation include:
• Inadequate fibre and/or fluid intake
• Psychological factors that can produce constipation through an inhibitory effect on the autonomic innervation of the colon. In some cases this may be related to poor toilet training during youth
• Poor liver function (bile is an excellent natural laxative).
Use of the well-known and much maligned anthraquinone-containing herbal laxatives is widespread. On balance, the evidence is that these herbs are safe and effective when used in the short term (see Chapter 2). However, they are best used as a last resort since their effect is only symptomatic. Their tendency to cause wind and griping can aggravate the pain associated with irritable bowel syndrome (IBS) and they are not at all suitable for constipation associated with bowel tension, spasm or irritability (see below). Also, the anthraquinone laxatives may become habit forming. This is for a number of reasons. One simple reason is that in overdose they empty a greater portion of the bowel than normal defaecation does. Hence, it is usual not to have a bowel movement the day following their use while this area of the bowel again fills. But the patient thinks that he or she is constipated again and will repeat the medication, thus perpetuating its use. The way around this is to take sufficient dose to cause a motion of normal consistency. This can vary greatly from person to person. This variability is because bowel flora activate the anthraquinones into their laxative form and bowel flora vary greatly from person to person.
The herbal treatment of constipation can be approached in the following way:
• Improve liver function with choleretic and cholagogue herbs, such as Chionanthus (fringe tree), Taraxacum (dandelion root), Silybum and Cynara
• Increase stool bulk through diet and with bulking herbs such as Ulmus (slippery elm) and Plantago ovata (ispaghula)
• Improve motor function with gastrointestinal spasmolytics such as Matricaria (chamomile) and Viburnum opulus (cramp bark)
• Improve gastrointestinal lubrication. Linseeds are particularly suitable because of their oil and mucilage content
• Judicious use of laxative herbs, beginning with gentle agents such as Juglans cinerea (butternut), Rumex (yellow dock), Glycyrrhiza (licorice) and Rehmannia. Otherwise a minimum quantity of Cassia (senna) or Rhamnus purshiana (cascara) can be introduced.
Rehmannia glutinosa | 1:2 | 25 mL |
Cynara scolymus | 1:2 | 20 mL |
Rumex crispus | 1:2 | 20 mL |
Taraxacum officinale root | 1:2 | 20 mL |
Matricaria recutita | 1:2 | 15 mL |
total | 100 mL |
Irritable bowel syndrome (IBS) is both a very common condition now attracting new research and a difficult disorder to comprehend and treat. One reason for this is that the diagnosis is only conclusively arrived at after exclusion of other known disorders. There are, however, more positive and integrated approaches available for managing this condition based on a broader psychosocial model,93 and phytotherapists are likely to find these more attractive. Possible aetiologic factors include hypersensitivity or motor dysfunction in the gut, abnormal response of smooth muscle to CNS signals, visceral and somatic sensory dysfunction and psychosocial distress.94
In addition to these ‘traditional’ factors involved in IBS, much research over the past decade or so has focussed on the pathogenic roles of altered enteric microbiota and inflammation. In addition to gastrointestinal pathogens, recent evidence suggests that patients with IBS have an abnormal composition and higher temporal instability of their intestinal flora.95 Because this flora is an important determinant of natural gut function and immunity, this instability is probably a contributing factor in IBS and suggests a role for specific management of intestinal dysbiosis in a subset of patients (see below). One specific and typical abnormal gut flora finding in IBS is small intestinal bacterial overgrowth.96 The fact that gastrointestinal infection or infestation, but not viral gastroenteritis, is a trigger of IBS in 10% of patients speaks to the role of disturbed microbial ecology in this disorder.97
On the topic of mucosal inflammation, one recent review of 16 studies found a relationship between mast cell abnormalities and symptom severity and frequency in IBS.98 The authors suggested that mast cell stabilisers warrant further assessment, suggesting a role for Albizia lebbek and Scutellaria baicalensis.
After years of inattention, there is a growing body of evidence that dietary components can contribute to IBS.99 While genuine food allergies are probably rare, food intolerances and especially malabsorption of particular sugars such as fructose (as treated by the so-called low FODMAP diet) do appear to be relevant in many patients. In addition, gluten restriction and sugar/carbohydrate restriction in general have all shown value in individual patients with IBS.99
A more definitive approach towards diagnosis and classification in IBS can lead to a better understanding and management. For example, it is unreasonable to assume that IBS characterised by diarrhoea would necessarily respond to the same treatment as IBS for which constipation predominates, yet many clinical trials make no attempt to evaluate therapy in terms of initial symptoms.
IBS is in fact the most common but yet least understood gastrointestinal disorder. There are three basic types:
• Functional diarrhoea with abdominal pain (diarrhoea-predominant IBS)
• Chronic abdominal pain and constipation (spastic colitis, or now termed constipation-predominant IBS)
• Abdominal pain with disturbed and variable bowel habit, typically constipation alternating with diarrhoea.
The dated term ‘mucous colitis’ refers to the excessive amount of mucus that can be passed with stools. As noted, the exact nature of the herbal treatment should depend on the factors identified in the individual case.
The first line of therapy for IBS is an increase in dietary fibre intake to improve intestinal transit time (especially for constipation-predominant IBS). If this fails, smooth muscle relaxants and/or antispasmodics are conventionally prescribed. A systematic review and meta-analysis to determine the efficacy of fibre intake, antispasmodics and peppermint oil as treatments for IBS identified 615 studies, but concluded that only 35 were eligible for inclusion.100 The beneficial effect of fibre compared with placebo or no treatment in 12 studies (n=591 patients) was limited to ispaghula (Plantago ovata) husk; wheat bran was no more effective than placebo or no treatment. Twelve different antispasmodics were evaluated in 22 studies compared with placebo or no treatment (n=1778 patients). Two antispasmodics (otilonium and hyoscine – an alkaloid from Hyoscyamus niger (henbane) and other Solanaceae plants) were deemed to be most efficacious. However, 14% of the patients who took antispasmodics reported adverse events, and more conservative strategies are worth attempting first.
The herbal remedy most researched for IBS is peppermint oil (see the peppermint monograph). Nine clinical trials on peppermint oil and its gastrointestinal effects, including a total of 269 patients, have been reviewed.101 Most studies of orally administered oil demonstrate a substantial spasmolytic effect on the gut, with slowed motility after prolonged use of doses above 0.2 mL. The authors concluded that an ideal formulation for the treatment of irritable bowel, and prevention of heartburn as a side effect through relaxation of the gastric sphincter, would be enteric coated and have a peak release at about 4 h after ingestion, with a release time of up to 24 h. Of 12 placebo-controlled trials reviewed in a subsequent paper,102 eight showed a statistically significant positive effect for peppermint oil over the placebo. The conclusion that peppermint oil ‘may be the drug of choice in IBS patients with non-serious constipation or diarrhoea to alleviate general symptoms and improve quality of life’ is, however, undermined by the wide range of placebo responses reported in the studies reviewed (between 10% and 52%). In a later rigorous study103 results indicated that 4 weeks of treatment with peppermint oil capsules was superior to a placebo in the treatment of IBS symptoms and its beneficial effect persisted 4 weeks after treatment had ended. This may be due to antibacterial actions of peppermint oil on enteric bacteria, or extended relaxant effects on smooth muscle tissue. Adverse event reports associated with peppermint oil were generally mild and transient and included heartburn and anal discomfort or burning. (See also the peppermint monograph.)
The need for enteric coating to demonstrate an effect for peppermint oil is a caution on the widespread recommendation for the use of other volatile antispasmodic constituents. The aromatic or pungent spices are, however, likely to add additional effects. A partially blinded, randomised, two-dose study on 207 subjects who had self-reported IBS symptoms for at least 3 months provided either 72 mg or 144 mg daily of standardised turmeric extract equivalent to 1800 or 3600 mg of dried root. The report showed overall benefits for both doses, though not clearly differentiated from background trends.104 Other spices particularly worth trying are Elettaria cardamomum (cardamom) and Foeniculum vulgare (fennel) and the subjective sensation of relief when taking remedies in this group is often striking.
Another approach is to use remedies with positive benefits on digestion higher up in the system. For example, about one-third of patients with functional dyspepsia also have IBS, and the symptoms of the two disorders overlap considerably: treatment for one might be applicable to the other. A postmarketing surveillance (uncontrolled) German study prospectively collected data over 6 weeks on 279 patients who reported having at least three of five IBS symptoms (abdominal pain, bloating, flatulence, right-sided abdominal cramps and constipation) and who took six capsules daily of a standardised extract of Cynara scolymus (artichoke) leaf extract equivalent to 320 mg per capsule. Improvement in their symptoms followed an average of 10 days of treatment (see also further studies in the globe artichoke monograph).105
All the above points to the value of trying some of the treatment options below:
• Spasmolytic herbs including Matricaria (chamomile), Humulus lupulus (hops), Viburnum opulus (cramp bark), Mentha piperita (peppermint) and any of the carminative spices
• Corydalis and Zingiber (ginger) may have a role in some patients through a modification of the perception of visceral organ pain
• Sedative and nervine tonic herbs, particularly Scutellaria lateriflora (skullcap) and Valeriana (valerian)
• Hepatorestorative and choleretic herbs to improve liver function such as Silybum (St Mary’s thistle), Cynara (globe artichoke leaf) and Schisandra
• Mucilage-containing herbs such as Ulmus (slippery elm), especially if there is constipation
• Gastrointestinal antiseptics to restore normal bowel flora such as Hydrastis (golden seal), propolis and oil of oregano. IBS patients may be intolerant of Allium (garlic), but if not it can be useful (see also under Intestinal dysbiosis below)
• The presence of mucus implies irritation, and gastrointestinal anti-inflammatories such as Filipendula (meadowsweet) and Matricaria (chamomile) are indicated
• Constipation should be treated with only gentle herbs such as Rumex crispus (yellow dock), Juglans cinerea (butternut) and Taraxacum (dandelion root)
• Symptoms of IBS are often confused or conflated with those of premenstrual and other hormonal upsets in women: in this case Vitex agnus-castus (chaste berry) and Trigonella foenum-graecum (fenugreek) are worth including
• An appropriate controlled exclusion diet should always be conducted to identify food irritants. These should however be done rigorously to avoid unnecessary elimination of nutritious food that has only occasional or apparent effect.
Case history
A female patient aged 42 complained of chronic episodes of discomfort and distension in the right lower abdomen. This could be quite sharp at times and was associated with a feeling of malaise. She was intolerant of fatty foods and had what she described as a ‘sluggish bowel’, although her motions were always ‘loose’. Past medical history revealed that she had lived on the Solomon Islands for an extended period during which time she had amoebic dysentery and malaria. A number of recent medical tests, including colonoscopy, could not find any abnormalities. She also had a history of heart damage of undefined origin. For this reason Crataegus (hawthorn) was included in her treatment (the Chinese use Crataegus as a digestive herb).
The following treatment was prescribed:
Crataegus spp. leaves | 1:2 | 20 mL |
Filipendula ulmaria | 1:2 | 20 mL |
Chionanthus virginicus | 1:2 | 15 mL |
Silybum marianum | 1:1 | 15 mL |
Matricaria chamomilla | 1:2 | 20 mL |
Viburnum opulus | 1:2 | 20 mL |
total | 110 mL |
Dose: 5 mL with water three times daily.
Ulmus (slippery elm), one heaped teaspoon with water three times a day, was also recommended. After 4 weeks she reported a stunning transformation. In the whole month the patient had only experienced one bad day. She was feeling very well, eating better and had more energy. The patient will remain on her herbal treatment for some time. This was a patient who had spent many years and thousands of dollars on conventional medical treatment and yet had remained unwell.
The most common causes of acute diarrhoea are infectious agents. Acute diarrhoea may also be caused by drugs or toxins. Chronic diarrhoea is also most likely to be caused by infectious agents. However, other common causes include inflammatory bowel diseases, malabsorption, IBS (idiopathic diarrhoea), medications and food additives. In all cases the source of the diarrhoea should be ascertained and appropriate treatment should then follow.
Acute gastrointestinal diarrhoea with vomiting is generally not suited to herbal therapy. This is because the patient will invariably vomit back the herbal treatment and may consequently develop an aversion to taking herbs.
Acute infectious diarrhoea in the absence of vomiting can be approached in the following way:
• Boost immunity with immune-enhancing herbs, particularly Echinacea and Andrographis
• Control fever with diaphoretic herbs such as Mentha piperita (peppermint) and Achillea (yarrow)
• If the infection does not involve a virus, Hydrastis (golden seal) or Berberis vulgaris (barberry) is indicated because of the antimicrobial activity of the berberine each contains (see the Berberis monograph). Berberine also inhibits the activity of enterotoxins. Other herbs rich in berberine could also be used such as Phellodendron and Coptis
• Antimicrobial essential oils safe for internal use can also be used to control gut infections. These include oils of anise, thyme and oregano. Allium (garlic) is also a useful, broad-spectrum gut antiseptic (raw crushed cloves or as the allicin-releasing powder)
• If cytotoxins or mucosal invasion are part of the pathogenic process, anti-inflammatory herbs such as Matricaria (chamomile) and mucilage-containing herbs such as Ulmus rubra (slippery elm) are indicated
• Tannin-containing herbs, such as Geranium maculatum (cranesbill), which act as astringents will also gently control diarrhoea without risk of aggravating the infection by reducing intestinal motility. They also reduce mucosal damage and are particularly indicated if the infection is due to a virus
• Antiprotozoal agents include propolis, Artemisia annua, berberine-containing herbs, Euphorbia and essential oil of oregano
• Normal conservative measures such as adequate fluid and electrolyte intake should also be implemented.
Chronic infectious diarrhoea is treated in a similar manner to acute infectious diarrhoea. However, particular emphasis should also be given to factors involved in host resistance:
• Gastrointestinal antiseptics (the essential oils noted above, Hydrastis), especially when used (but not concurrently) with agents that encourage growth of normal flora, will help to restore the protective activity of intestinal flora (see below)
• Herbs to improve gastric acidity to prevent reinfection may also need emphasis. These include Coleus, Angelica, Zingiber (ginger), Capsicum (cayenne) and bitters such as Gentiana.
Case history
A female patient aged 35 presented with chronic infection with the protozoan Giardia that had persisted for more than 3 months. She was prescribed the following treatment (based on 1 week):
Echinacea angustifolia root | 1:2 | 35 mL |
Picrorrhiza kurroa | 1:2 | 10 mL |
Angelica archangelica | 1:2 | 15 mL |
Propolis | 1:10 | 20 mL |
Zingiber officinale | 1:2 | 5 mL |
Matricaria chamomilla | 1:2 | 15 mL |
total | 100 mL |
Dose: 5 mL with water three times a day.
Hydrastis (golden seal) 500 mg tablets at 4/day were also prescribed and regular intake of a Lactobacillus culture at separate times was recommended.
After 4 weeks symptoms were about the same. There was even a 1-week period when the patient felt that the herbs were aggravating her condition. After another 4 weeks there was considerable improvement and the condition was resolved by a further 8 weeks’ treatment. Note that Picrorrhiza in the formula doubled as a bitter to increase gastric acid and as an immune-enhancing agent and could be substituted with Andrographis, which has similar properties.
Diverticulosis describes the presence of diverticula or pockets in the wall of the large bowel. Acute diverticulitis is the clinical syndrome that occurs when a diverticulum becomes inflamed and perforates, which is relatively uncommon.
Lack of dietary fibre is probably the main cause of diverticulosis, but other factors that lead to constipation and colonic hypermobility may be involved. (Colonic hypermobility results in excessive mixing activity, which exacerbates constipation and causes areas of raised intraluminal pressure.)
Excessive pressure in the colon associated with age-related weakness and stiffness of the colon wall are probably all involved. If faecal volume is habitually small, pressure may rise to excessive levels during segmenting (mixing) movements. This can result in ‘blow outs’, which are herniations of the mucosa at naturally weak places in the wall where arteries pass inwards. With age, these can develop into diverticula. Diverticula are potential areas of stagnation in the bowel because they have no muscular wall and, therefore, are less subject to flushing by normal mixing movements.
The main aim of herbal treatment in uncomplicated diverticular disease is to reduce stagnation and further degeneration of the bowel wall. Aspects of treatment include the following:
• Appropriate dietary measures to increase fibre intake (but excluding seeds and nuts) and supplementation with mucilaginous herbs such as Ulmus (slippery elm). This will also help to maintain healthy bowel flora
• Gastrointestinal spasmolytics to decrease intracolonic pressure, including Viburnum opulus (cramp bark), Dioscorea (wild yam) and Matricaria (chamomile)
• Herbs to improve connective tissue strength including herbs containing flavonoids and oligomeric procyanidins (OPCs) such as Vitis (grape seed extract) and Crataegus (hawthorn). Polygonum multiflorum is also thought to improve connective tissue and is also a gentle laxative
• Gentle treatment of any associated constipation (see above).
Painful or symptomatic diverticular disease can also occur in the absence of diverticulitis. In medical thinking this is considered to be a variant of irritable bowel syndrome. However, it could result from a low-grade ‘diverticulosis’. Depending on the assessment of the patient, this problem should either be treated as irritable bowel syndrome or the treatment approach described below to prevent recurrence of acute diverticulitis should otherwise be followed.
Acute diverticulitis usually requires hospitalisation. Herbal treatment is more suited to prevention of its recurrence. As well as incorporating the aspects of treatment of uncomplicated diverticular disease described above, the approach to prevention of acute diverticulitis should additionally include:
• Immune-enhancing herbs such as Echinacea and Andrographis to control pathogenic bacteria
• Gastrointestinal antiseptic herbs (see Gastrointestinal infections above)
• Anti-inflammatory gastrointestinal herbs such as Filipendula (meadowsweet) and Matricaria (chamomile).
Case history
A male patient aged 72 suffered an attack of acute diverticulitis and was concerned to prevent another episode.
Treatment consisted of the following prescription (based on 1 week):
Echinacea angustifolia root | 1:2 | 25 mL |
Hydrastis canadensis | 1:3 | 20 mL |
Viburnum opulus | 1:2 | 20 mL |
Matricaria recutita | 1:2 | 20 mL |
Propolis | 1:10 | 15 mL |
total | 100 mL |
Dose: 5 mL with water three times a day.
Ulmus (slippery elm) powder, one heaped teaspoon with water before each meal, was also prescribed.
The patient was also advised to have more fibre in his diet, particularly more fruit. Fresh crushed garlic, one to two cloves a day, was also recommended for 3 days of every week. After 6 months the herbal treatment was discontinued but the fresh garlic (for 1 to 2 days/week), slippery elm and dietary changes were still observed. Several years later, the patient had not experienced any recurrence of acute diverticulitis.
As well as the obvious involvement of pathogenic gut micro-organisms in acute and chronic gut infections, they are probably contributing factors in the chronicity of disturbances of immune or gut function such as food sensitivity, IBS, asthma, dermatitis, psoriasis and autoimmune diseases such as rheumatoid arthritis, inflammatory bowel disease and ankylosing spondylitis.
The involvement of gut flora in autoimmune disease is exemplified by published studies suggesting the involvement of pathogenic organisms in several autoimmune diseases. Dr Alan Ebringer of King’s College Hospital, London, decided to test this association clinically. Patients with ankylosing spondylitis were placed on a low starch diet, because it was postulated that this would reduce the number of Klebsiella organisms in the gut.106 Most patients on this programme had their disease process halted, but the diet must be adhered to for at least 6 months.
In addition, many people who are chronically unwell but with no specific diagnosis may be suffering from a microbial dysbiosis, which is not sufficiently marked to be classified as an infection. This is well illustrated by the postulated involvement of Candida albicans in many such syndromes.
The gastric acid barrier is an important factor in maintaining healthy gut microflora, and other digestive secretions such as bile and pancreatic enzymes also play a role. Immune function, especially secretory IgA, diet and bowel motility can also influence the location and levels of particular microflora.
The assumption made after episodes of gut infection is that recovery is complete. However, several pathogenic microorganisms can take up residence in the gut at low levels following infection. The implication of this can be chronic disease. The role of antibiotics, especially their chronic use, in contributing to gut flora dysbiosis is widely recognised.
The suggested programme below has been adapted from the approach developed by the late Hein Zeylstra (to whom this book is dedicated), for the management of inflammatory bowel disease. It can work well in conjunction with a low starch diet, as per Ebringer above.
Herbal treatment for gut flora dysbiosis is compatible with the use of probiotics and prebiotics. In fact the protocol below relies on the use of a herbal ‘prebiotic’. If probiotics are additionally prescribed (either via supplements or diet), then they should not be taken at the same time as antimicrobial herbs:
• The herbal approach is based on the ‘weed’ and ‘feed’ hypothesis. In other words, the aim is to manipulate existing microflora to provide a healthier balance and in particular create a predominance of organisms (such as Lactobacilli and Bifidobacteria) that do not imbalance the immune system. By manipulating existing resident microflora, the changes in gut flora are more likely to be permanent
• The ‘weed-killer’ consists of using broad-spectrum antimicrobial herbs that have activity against bacteria, fungi and protozoa. By using such agents, levels of most microflora are depressed and there is less likelihood of exacerbating the gut flora imbalance. The ‘fertiliser’ then consists of agents such as marshmallow root glycetract, aloe vera concentrate or slippery elm powder that selectively encourage healthy microflora, along the same principles as the use of prebiotics
• This process is repeated over weekly cycles. For 2 to 3 days only of each week (preferably the weekend) the antimicrobial herbs are taken throughout the day. Then the ‘fertiliser’ herbs are used for the rest of the week. The cycle is then repeated for at least 6 weeks, although much longer may be required, especially in the case of chronic diseases such as autoimmune disease
• Key broad-spectrum antimicrobial herbs are garlic, berberine-containing herbs (such as Hydrastis or Phellodendron), essential oils (e.g. oregano) and mild tannin-containing herbs (especially green tea and grape seed extracts). The advantage of tannins is that they are poorly absorbed in the gastrointestinal tract and are carried to the large bowel. Through their capacity to bind proteins, they can inhibit the growth of all microorganisms, but appear to be specifically selective on pathogenic organisms (see below).
• The antimicrobial herbs need to be taken in reasonably high doses on the 2 to 3 ‘weeding’ days
• However, the green tea and grape seed extracts are best taken only during the ‘feeding’ days as they will have minimal impact on healthy flora, but will provide a dampening effect on the regrowth of pathogenic bacteria.
Grape seed oligomeric and polymeric procyanidins demonstrated a beneficial effect on caecal fermentation in rats. Caecal pH decreased, and fermentative activity was stimulated without an increase of deleterious enzymatic activity.107 A small clinical study in Japan demonstrated that a green tea preparation was able to positively affect intestinal dysbiosis in nursing home patients by raising levels of lactobacilli and bifidobacteria, lowering levels of Enterobacteriaceae, Bacteroidaceae and Eubacteria, and decreasing odorous compounds. Levels of pathogenic bacterial metabolites were also decreased.108,109 A further study found that supplementation with tea catechins produced favourable improvements in the participant’s bowel condition, as evidenced by a reduction in faecal moisture, pH, ammonia, sulphide and oxidation-reduction potential. In both trials the dose was 300 mg/day of tea catechins, which is equivalent to about six cups of green tea.110
In summary, on Saturday and Sunday (and perhaps Monday) the patient is advised to take antimicrobial herbs in high doses spread throughout the day. These would include Allium (garlic), oil of oregano and a source of berberine such as Phellodendron. During the rest of the week Ulmus (slippery elm), green tea and grape seed extract are advised. The cycle is then repeated for at least 6 weeks.
The principles of treating chronic inflammatory conditions have been rehearsed in the Inflammatory and autoimmune diseases section in Chapter 8. Inflammatory bowel diseases (IBDs), like Crohn’s disease and ulcerative colitis, however, present local digestive challenges that also deserve separate discussion.
Clearly the main focus in a systematic approach to the digestive role in IBD is to consider diet as a factor. The potential link between Crohn’s and wheat consumption has been referred to in Chapter 8 and these and other possibilities are worth rigorously exploring. A low sulphur diet for ulcerative colitis111 and a dairy- and yeast-free diet for Crohn’s disease are also worth trialling.
There is a potential role for topical herbal treatments. Aloe vera gel is widely promoted for the treatment of such digestive disorders. In a randomised, placebo-controlled, blinded study 100 mL of Aloe barbadensis (Aloe vera) gel twice daily was shown to have modest benefits in the treatment of mild to moderate ulcerative colitis, though only on subjective symptoms.112
IBDs are marked by increased production of pro-inflammatory cytokines such as tumour necrosis factor (TNF)-alpha, interleukin-1 (IL-1) and IL-6. There is the theoretical possibility that reduction in these markers may be obtained by remedies acting primarily on the digestive system. In a randomised, open label, multi-centre trial, 20 patients with active Crohn’s receiving standard treatment excluding TNF-alpha inhibitors ingested 250 mg standardised capsules containing wormwood (Artemisia absinthium, absinthin at 0.32% to 0.38%) with Elettaria cardamomum (cardamom) seeds and Pistacia lentiscus (mastic), three times a day. These herbs might all be chosen as treatments for inflammation of the bowel. Blood samples were tested at 3 and 6 weeks to determine TNF-alpha levels compared with baseline and these were significantly lowered in the treatment group compared with placebo.113
The important prospect of treating IBD with Boswellia has been commented on in the Boswellia monograph.
It is known that bile can be a co-factor in IBD, particularly following secondary metabolism by bowel flora. Traditional phytotherapeutic approaches have included remedies that are known to have choleretic or hepatic effects and these can be seen as plausible strategies. Disturbed bowel flora may have additional harmful effects and steps to reduce dysbiosis are often justified.
Taking these approaches into account, with the obvious caution of viewing IBD as a systemic disorder as much as a local one, the following measures might be considered. Reference should be made also to the section above on Intestinal dysbiosis for other important therapeutic considerations:
• Immune modulating herbs such as Echinacea root are useful and appear to be able to regulate the inflammation (see the Echinacea monograph for a discussion of its role in autoimmune disease)
• Anti-inflammatory herbs especially Boswellia in solid extract form, as well as Calendula officinalis (marigold) and Curcuma longa (turmeric) can be helpful (see the Boswellia and turmeric monographs for relevant clinical data)
• As with diverticular disease, increased ‘soft’ fibre intake and mucilaginous plants and materials, such as aloe gel is likely to be helpful. Mucilaginous gums and prebiotic constituents will also help to maintain healthy bowel flora and counteract negative effects of bile irritation. Those with additional inflammatory-modulating effects such as Linum usitassimum (flaxseed) or Trigonella foenum-graecum (fenugreek) may be particularly helpful
• Choleretic and hepatic remedies such as Cynara scolymus (globe artichoke), Silybum marianum (St Mary’s thistle), Hydrastis canadensis (golden seal) and Berberis vulgaris (barberry) are often useful in reducing deleterious effects of the ‘wrong sort of bile’.
Case history
A 39-year-old woman had suffered from Crohn’s disease for 24 years. Lately (the last 2.5 years) it had been out of control. She had a stricture 5 cm from the rectum which made it difficult to control her bowel motions. Her main symptoms were almost continuous diarrhoea. Other symptoms included mouth ulcers, occasional high temperatures and chronically blocked sinuses. Conventional medications were mesalazine 1500 mg/day, oral prednisone (tapering off) and prednisone enemas daily. The following herbal treatments were instituted:
• The intestinal dysbiosis protocol above for 6 weeks and then Hydrastis (golden seal) 1500 mg/day and Ulmus (slippery elm) powder on a regular basis (she could not tolerate garlic)
• Tablets containing Echinacea angustifolia root (600 mg) and Echinacea purpurea root (675 mg) (3 per day)
• Tablets containing Boswellia (1200 mg), turmeric (2000 mg), celery (1000 mg) and ginger (300 mg) (4 per day)
After 5 month’s treatment there was great symptomatic improvement and the patient commented: ‘I no longer view travelling in terms of toilet stops’. She had one relatively normal bowel motion per day, no mouth ulcers and her high temperatures were gone. After 12 months there was no need for the prednisone enemas. After 36 months the patient was completely asymptomatic and on minimal conventional medication. Her lifestyle was transformed!
A haemorrhoid or a pile is a dilation of the internal haemorrhoidal plexus. Internal haemorrhoids are more significant and are covered with mucous membrane. An external pile is really a rupture of a small vein in the perianal region. The exact cause is not known, but is probably associated with a mild partial mucosal prolapse during straining at defaecation.
Haemorrhoids are aggravated by pelvic congestion (constipation and pregnancy) and prostatic enlargement. Some degree of loss of elasticity of the anal sphincter may play a role. Lifting heavy weights may aggravate the condition. Treatment could include the following:
• Increase dietary fibre, both soluble and insoluble
• Mucilage-containing herbs such as Ulmus (slippery elm) and psyllium to keep the stool soft
• Any associated constipation should be treated (see above)
• Oral treatment using herbs to improve venous and connective tissue tone. These include Aesculus (horsechestnut), Ruscus (butcher’s broom) and Polygonum multiflorum. Flavonoid-containing herbs, such as Crataegus, also have this property. Melilotus (sweet clover) helps to relieve tissue congestion
• Topical treatment with healing and astringent herbs such as Hamamelis (witchhazel), Symphytum (comfrey) and Calendula. Aesculus also works well topically, especially in gel formulations, but should not be applied if the piles are bleeding (see monograph)
• If liver congestion exists, which will exacerbate pelvic congestion, treatment with choleretic and hepatoprotective herbs should be applied.
Case history
A male patient aged 35 had suffered from haemorrhoids for 8 years. He had been treated with rubber ligation but was still suffering problems such as irritation and occasional bleeding. He was not experiencing constipation but felt tense in the lower abdomen and was generally an anxious person. Other symptoms included indigestion, abdominal bloating and reflux.
Treatment consisted of (based on 1 week):
Artemisia absinthium | 1:5 | 10 mL |
Aesculus hippocastanum | 1:2 | 25 mL |
Melilotus officinale | 1:2 | 20 mL |
Ruscus aculeatus | 1:2 | 25 mL |
Valeriana officinalis | 1:2 | 20 mL |
total | 100 mL |
Dose: 5 mL with water three times daily.
Ulmus (slippery elm) powder, one heaped teaspoon twice a day was also prescribed, together with comfrey ointment.
Over the course of the next 16 weeks, the condition improved and he was free of any symptoms related to the haemorrhoids. He also reported feeling more relaxed.
References
1. Du P, O’Grady G, Davidson JB, et al. Multiscale modeling of gastrointestinal electrophysiology and experimental validation. Crit Rev Biomed Eng. 2010;38(3):225–254.
2. Altaf MA, Sood MR. The nervous system and gastrointestinal function. Dev Disabil Res Rev. 2008;14(2):87–95.
3. Waterman SA, Tonini M, Costa M. The role of ascending excitatory and descending inhibitory pathways in peristalsis in the isolated guinea-pig small intestine. J Physiol (Lond). 1994;481(pt 1):223–232.
4. De Ponti F, Cosentino M, Lecchini S, et al. Physiopharmacology of the peristaltic reflex: an update. Ital J Gastroenterol. 1991;23(5):264–269.
5. Wood JD. Enteric nervous system: sensory physiology, diarrhea and constipation. Curr Opin Gastroenterol. 2010;26(2):102–108.
6. Wingate DL. The effect of diet on small intestinal and biliary tract function. Am J Clin Nutr. 1985;42(5 suppl):1020–1024.
7. Hoogerwerf WA. Role of biological rhythms in gastrointestinal health and disease. Rev Endocr Metab Disord. 2009;10(4):293–300.
8. Mellander A, Abrahamsson H, Sjovall H. The migrating motor complex – the motor component of a cholinergic enteric secretomotor programme? Acta Physiol Scand. 1995;154(3):329–341.
9. May B, Kuntz H, Kieser M, Koler S. Efficacy of a fixed peppermint oil/caraway oil combination in non-ulcer dyspepsia. Arzneimittelforschung. 1996;46(II):1149–1153.
10. De Winter BY, De Man JG. Interplay between inflammation, immune system and neuronal pathways: effect on gastrointestinal motility. World J Gastroenterol. 2010;16(44):5523–5535.
11. Cooke HJ. Neuroimmune signaling in regulation of intestinal ion transport. Am J Physiol. 1994;266(2 pt 1):167–178.
12. Santos J, Alonso C, Vicario M, et al. Neuropharmacology of stress-induced mucosal inflammation: implications for inflammatory bowel disease and irritable bowel syndrome. Curr Mol Med. 2008;8(4):258–273.
13. Wouters MM, Boeckxstaens GE. Neuroimmune mechanisms in functional bowel disorders. Neth J Med. 2011;69(2):55–61.
14. Snoek SA, Verstege MI, Boeckxstaens GE. The enteric nervous system as a regulator of intestinal epithelial barrier function in health and disease. Expert Rev Gastroenterol Hepatol. 2010;4(5):637–651.
15. Surawicz CM. Mechanisms of diarrhea. Curr Gastroenterol Rep. 2010;12(4):236–244.
16. Ralph A, Giannella MD, Selwyn A, et al. Influence of gastric acidity on bacterial and parasitic enteric infections. Ann Intern Med. 1973;78(2):271–276.
17. Torsoli A, Severi C. The neuroendocrine control of gastrointestinal motor activity. J Physiol (Paris). 1993;87(6):367–374.
18. Mach T. The brain-gut axis in irritable bowel syndrome – clinical aspects. Med Sci Monit. 2004;10(6):125–131.
19. Chen CL, Liu TT, Yi CH, Orr WC. Effects of capsaicin-containing red pepper sauce suspension on esophageal secondary peristalsis in humans. Neurogastroenterol Motil. 2010;22(11):1177–1182.
20. Szolcsányi J. Forty years in capsaicin research for sensory pharmacology and physiology. Neuropeptides. 2004;38(6):377–384.
21. Serdiuk SE, Komissarov IV, Gmiro VE. The role of the chemosensory systems in the inhibitory regulation of cholinergic transmission in the small intestine. Fiziol Zh. 1993;39(1):54–61.
22. Furness JB, Costa M. Projections of intestinal neurons showing immunoreactivity for vasoactive intestinal polypeptide are consistent with these neurons being the enteric inhibitory neurons. Neurosci Lett. 1979;15(2–3):199–204.
23. Donnerer J, Bartho L, Holzer P, Lembeck F. Intestinal peristalsis associated with release of immunoreactive substance P. Neuroscience. 1984;11(4):913–918.
24. Glatzel H, Rüberg-Schweer M. Regional influence on cutaneous blood flow effected by oral spice intake. Nutr Dieta Eur Rev Nutr Diet. 1968;10:194–214.
25. Staljanssens D, Azari EK, Christiaens O. The CCK(-like) receptor in the animal kingdom: functions, evolution and structures. Peptides. 2011;32(3):607–619.
26. Bradwejn J, Zhou Y, Koszycki D, Shlik J. A double blind, placebo-controlled study on the effects of Gotu Kola (Centella asiatica) on acoustic startle response in healthy subjects. J Clin Psychopharmacol. 2000;20(6):680–684.
27. Eckel LA, Ossenkopp KP. Cholecystokinin reduces sucrose palatability in rats: evidence in support of a satiety effect. Am J Physiol. 1994;267(6 pt 2):R1496–R1502.
28. Holt S, Brand J, Soveny C, Hansky J. Relationship of satiety to postprandial glycaemic, insulin and cholecystokinin responses. Appetite. 1992;18(2):129–141.
29. Hansen JB, Arkhammar PO, Bodvarsdottir TB, Wahl P. Inhibition of insulin secretion as a new drug target in the treatment of metabolic disorders. Curr Med Chem. 2004;11(12):1595–1615.
30. Bartness TJ, Waldbillig RJ. Cholecystokinin-induced suppression of feeding: an evaluation of the generality of gustatory-cholecystokinin interactions. Physiol Behav. 1984;32(3):409–415.
31. Morley JE, Silver AJ. Anorexia in the elderly. Neurobiol Aging. 1988;9(1):9–16.
32. Khayyal MT, Seif-El-Nasr M, El-Ghazaly MA, et al. Mechanisms involved in the gastro-protective effect of STW 5 (Iberogast) and its components against ulcers and rebound acidity. Phytomedicine. 2006;13(suppl 5):56–66.
33. Takeuchi T, Shiratori K, Watanabe S, et al. Secretin as a potential mediator of antiulcer actions of mucosal protective agents. J Clin Gastroenterol. 1991;13(suppl):S83–S87.
34. Shiratori K, Watanabe S, Takeuchi T. Effect of licorice extract (Fm100) on release of secretin and exocrine pancreatic secretion in humans. Pancreas. 1986;1(6):483–487.
35. Minton NA. Volunteer models for predicting antiemetic activity of 5-HT3-receptor antagonists. Br J Clin Pharmacol. 1994;37(6):525–530.
36. Costall B, Naylor RJ. Neuropharmacology of emesis in relation to clinical response. Br J Cancer. 1992;19:2–7.
37. Beubler E, Schirgi-Degen A. Serotonin antagonists inhibit sennoside-induced fluid secretion and diarrhea. Pharmacology. 1993;47(suppl):64–69.
38. Hyland NP, Cryan JF. Gut feeling about GABA: focus on GABA(B) receptors. Front Pharmacol. 2010;1:124.
39. Krantis A, Costa M, Furness JB, Orbach J. Gamma-aminobutyric acid stimulates intrinsic inhibitory and excitatory nerves in the guinea-pig intestine. Eur J Pharmacol. 1980;67(4):461–468.
40. Izzo AA, Sharkey KA. Cannabinoids and the gut: new developments and emerging concepts. Pharmacol Ther. 2010;126(1):21–38.
41. Korczynski W, Ceregrzyn M, Matyjek R, et al. Central and local (enteric) action of orexins. J Physiol Pharmacol. 2006;57(suppl 6):17–42.
42. Bjarnason I. Intestinal permeability. Gut. 1994;35(suppl 1):18–22.
43. Lee EB, Kim OK, Jung CS, Jung KH. The influence of methanol extract of Ulmus davidiana var. Japonica cortex on gastric erosion and ulcer and paw edema in rats. Yakhak Hoeji. 1995;39(6):671–675.
44. Blaut M, Collins MD, Welling GW, et al. Molecular biological methods for studying the gut microbiota: the EU human gut flora project. Br J Nutr. 2002;87(suppl 2):203–211.
45. Van Eldere J, Robben J, Caenepeel P, Eyssen H. Influence of a cecal volume-reducing intestinal microflora on the excretion and entero-hepatic circulation of steroids and bile acids. J Steroid Biochem. 1988;29(1):33–39.
46. Ozawa A, Ohnishi N, Tazume S, et al. Intestinal bacterial flora and host defense mechanisms. Tokai J Exp Clin Med. 1986;11(suppl):65–79.
47. Rolfe RD. Interactions among microorganisms of the indigenous intestinal flora and their influence on the host. Rev Infect Dis. 1984;6(suppl 1):S73–S79.
48. Walker WA, Bloch KJ. Gastrointestinal transport of macromolecules in the pathogenesis of food allergy. Ann Allergy. 1983;51(2 pt 2):240–245.
49. Turnbaugh PJ, Ridaura VK, Faith JJ, et al. The effect of diet on the human gut microbiome. Sci Transl Med. 2009;1(6):6–14.
50. De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA. 2010;107:14691–14696.
51. Sullivan A, Edlund C, Nord CE. Effect of antimicrobial agents on the ecological balance of human microflora. Lancet Infect Dis. 2001;1:101–114.
52. Wickens K, Pearce N, Crane J, Beasley R. Antibiotic use in early childhood and the development of asthma. Clin Exp Allergy. 1999;29:766–771.
53. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? Trends Microbiol. 2004;12:562–568.
54. Parsonnet J. Bacterial infection as a cause of cancer. Environ Health Perspect. 1995;103(suppl 8):263–268.
55. Tazume S, Ozawa A, Yamamoto T, et al. Ecological study on the intestinal bacteria flora of patients with diarrhea. Clin Infect Dis. 1993;16(suppl 2):77–82.
56. Gorbach SL. Estrogens, breast cancer, and intestinal flora. Rev Infect Dis. 1984;6(suppl 1):85–90.
57. Sacquet E, Leprince C, Riottot M, Raibaud P. Dietary fiber and cholesterol and bile acid metabolism in axenic (germfree) and holoxenic (conventional) rats. III. Effect of nonsterilized pectin. Reprod Nutr Dev. 1985;25(1A):93–100.
58. Sherman PM, Ossa JC, Johnson-Henry K. Unraveling mechanisms of action of probiotics. Nutr Clin Pract. 2009;24(1):10–14.
59. Roberfroid M, Gibson GR, Hoyles L, et al. Prebiotic effects: metabolic and health benefits. Br J Nutr. 2010;104(suppl 2):1–63.
60. Broekaert WF, Courtin CM, Verbeke K, et al. Prebiotic and other health-related effects of cereal-derived arabinoxylans, arabinoxylan-oligosaccharides, and xylooligosaccharides. Crit Rev Food Sci Nutr. 2011;51(2):178–194.
61. Praznik W, Spies T. Fructo-oligosaccharides from Urginea maritima. Carbohydr Res. 1993;243(1):91–97.
62. Mitsuoka T, Hidaka H, Eida T. Effect of fructo-oligosaccharides on intestinal microflora. Nahrung. 1987;31(5–6):427–436.
63. Bouhnik Y, Flourié B, Riottot M, et al. Effects of fructooligosaccharides ingestion on fecal bifidobacteria and selected metabolic indexes of colon carcinogenesis in healthy humans. Nutr Cancer. 1996;26(1):21–29.
64. Saetta JP, March S, Gaunt ME, Quinton DN. Gastric emptying procedures in the self-poisoned patients: are we forcing gastric content beyond the pylorus? J R Soc Med. 1991;84(5):274–276.
65. Vale JA. Primary decontamination: vomiting, gastric irrigation or only medicinal charcoal? Ther Umsch. 1992;49(2):102–106.
66. Underhill TJ, Greene MK, Dove AF. A comparison of the efficacy of gastric lavage, ipecacuanha and activated charcoal in the emergency management of paracetamol overdose. Arch Emerg Med. 1990;7(3):148–154.
67. Ilett KF, Gibb SM, Unsworth RW. Syrup of ipecacuanha as an emetic in adults. Med J Aust. 1997;2(3):91–93.
68. Ramberg JE, Nelson ED, Sinnott RA. Immunomodulatory dietary polysaccharides: a systematic review of the literature. Nutr J. 2010;9:54.
69. Chattopadhyay A, Shetty KV. Recurrent aphthous stomatitis. Otolaryngol Clin North Am. 2011;44(1):79–88.
70. Burgess JA, van der Ven PF, Martin M, et al. Review of over-the-counter treatments for aphthous ulceration and results from use of a dissolving oral patch containing Glycyrrhiza complex herbal extract. J Contemp Dent Pract. 2008;9(3):88–98.
71. Roesch W, Liebregts T, Gundermann KJ, et al. Phytotherapy for functional dyspepsia: a review of the clinical evidence for the herbal preparation STW 5. Phytomedicine. 2006;13(suppl 5):114–121.
72. Milzer J, Iten F, Reichling J, Sallre R. Iberis amara L. and Iberograst – results of a systematic review concerning functional dyspepsia. J Herbal Pharmacother. 2004;4(4):51–59.
73. Wegener T, Wagner H. The active components and the pharmacological multi-target principle of STW 5 (Iberogast). Phytomedicine. 2006;13(suppl 5):20–35.
74. Rohof WO, Hirsch DP, Boeckxstaens GE. Pathophysiology and management of gastroesophageal reflux disease. Minerva Gastroenterol Dietol. 2009;55(3):289–300.
75. Saarela R, Lindroos E, Soini H, et al. Chewing problems and mortality. J Am Geriatr Soc. 2011;59(1):181–183.
76. Northridge ME, Nye A, Zhang YV, et al. ‘Third places’ for healthy aging: online opportunities for health promotion and disease management in adults in Harlem. J Am Geriatr Soc. 2011;59(1):175–176.
77. Anand G, Katz PO. Gastroesophageal reflux disease and obesity. Gastroenterol Clin North Am. 2010;39(1):39–46.
78. Graham DY, Anderson S, Lang T. Garlic or Jalapeno peppers for treatment of Helicobacter pylori infection. Am J Gastrol. 1999;94(5):1200–1202.
79. Yarnell E, Abascal K. Antiadhesion herbs. Altern Complement Ther. 2008;14(3):139–144.
80. Misciagna G, Cisternino AM, Freudenheim J. Diet and duodenal ulcer. Dig Liver Dis. 2000;32(6):468–472.
81. Calam J, Baron JH. ABC of the upper gastrointestinal tract: pathophysiology of duodenal and gastric ulcer and gastric cancer. BMJ. 2001;323(7319):980–982.
82. Suadicani P, Hein HO, Gyntelberg F. Genetic and life-style determinants of peptic ulcer. A study of 3387 men aged 54 to 74 years: The Copenhagen Male Study. Scand J Gastroenterol. 1999;34(1):12–17.
83. Rosenstock S, Jørgensen T, Bonnevie O, Andersen L. Risk factors for peptic ulcer disease: a population based prospective cohort study comprising 2416 Danish adults. Gut. 2003;52(2):186–193.
84. Parasher G, Eastwood GL. Smoking and peptic ulcer in the Helicobacter pylori era. Eur J Gastroenterol Hepatol. 2000;12(8):843–853.
85. Eastwood GL. Is smoking still important in the pathogenesis of peptic ulcer disease? J Clin Gastroenterol. 1997;25(suppl 1):S1–S7.
86. Levenstein S. Peptic ulcer at the end of the 20th century: biological and psychological risk factors. Can J Gastroenterol. 1999;13(9):753–759.
87. Melmed RN, Gelpin Y. Duodenal ulcer: the helicobacterization of a psychosomatic disease? Isr J Med Sci. 1996;32(3–4):211–216.
88. Abdel-Salam OM, Czimmer J, Debreceni A, et al. Gastric mucosal integrity: gastric mucosal blood flow and microcirculation. An overview. J Physiol (Paris). 2001;95(1–6):105–127.
89. Bandyopadhyay D, Biswas K, Bhattacharyya M, et al. Gastric toxicity and mucosal ulceration induced by oxygen-derived reactive species: protection by melatonin. Curr Mol Med. 2001;1(4):501–513.
90. Majumdar AP, Fligiel SE, Jaszewski R. Gastric mucosal injury and repair: effect of aging. Histol Histopathol. 1997;12(2):491–501.
91. Al Mofleh IA. Spices, herbal xenobiotics and the stomach: friends or foes? World J Gastroenterol. 2010;16(22):2710–2719.
92. Roberts F. Modern Herbalism For Digestive Disorders. Northamptonshire: Thomsons, 1981.
93. Tanaka Y, Kanazawa M, Fukudo S, Drossman DA. Biopsychosocial model of irritable bowel syndrome. J Neurogastroenterol Motil. 2011;17(2):131–139.
94. Hasler WL. Traditional thoughts on the pathophysiology of irritable bowel syndrome. Gastroenterol Clin North Am. 2011;40(1):21–43.
95. Bolino CM, Bercik P. Pathogenic factors involved in the development of irritable bowel syndrome: focus on a microbial role. Infect Dis Clin North Am. 2010;24(4):961–975.
96. Yamini D, Pimentel M. Irritable bowel syndrome and small intestinal bacterial overgrowth. J Clin Gastroenterol. 2010;44(10):672–675.
97. Spiller R, Garsed K. Postinfectious irritable bowel syndrome. Gastroenterology. 2009;136(6):1979–1988.
98. Ford AC, Talley NJ. Mucosal inflammation as a potential etiological factor in irritable bowel syndrome: a systematic review. J Gastroenterol. 2011;46(4):421–431.
99. Eswaran S, Tack J, Chey WD. Food: the forgotten factor in the irritable bowel syndrome. Gastroenterol Clin North Am. 2011;40(1):141–162.
100. Ford AC, Talley NJ, Spiegel BMR, et al. Effect of fibre, antispasmodics, and peppermint oil in the treatment of irritable bowel syndrome: systematic review and meta-analysis. BMJ. 2008;337:a2313.
101. Grigoleit HG, Grigoleit P. Gastrointestinal clinical pharmacology of peppermint oil. Phytomedicine. 2005;12:607–611.
102. Grigoleit HG, Grigoleit P. Peppermint oil in irritable bowel syndrome. Phytomedicine. 2005;12:601–606.
103. Cappello G, Spezzaferro M, Grossi L, et al. Peppermint oil (Mintoil[r]) in the treatment of irritable bowel syndrome: a prospective double blind placebo-controlled randomized trial. Dig Liver Dis. 2007;39(6):530–536.
104. Bundy R, Walker A, Middleton R, Booth J. Turmeric extract may improve irritable bowel syndrome symptomology in otherwise healthy adults: pilot study. J Altern Complement Med. 2004;10(6):1015–1018.
105. Walker AF, Middleton RW, Petrowicz O. Artichoke leaf extract reduces symptoms of irritable bowel syndrome in a post-marketing surveillance study. Phytother Res. 2001;15:58–61.
106. Ebringer A, Wilson C. The use of a low starch diet in the treatment of patients suffering from ankylosing spondylitis. Clin Rheumatol. 1996;15(suppl 1):62–66.
107. Tebib K, Besancon P, Rouanet JM. Effects of dietary grape seed tannins on rat cecal fermentation and colonic bacterial enzymes. Nutr Res. 1996;16(1):105–110.
108. Hara Y. Influence of tea catechins on the digestive tract. J Cell Biochem Suppl. 1997;27:52–58.
109. Goto K, Kanaya S, Nishikawa T, et al. Green tea catechins improve gut flora. Ann Long-Term Care. 1998;6:1–7.
110. Goto K, Kanaya S, Ishigami T, Hara Y. The effects of tea catechins on fecal conditions of elderly residents in a long-term care facility. J Nutr Sci Vitaminol. 1999;45(1):135–141.
111. Roediger WE. Decreased sulphur aminoacid intake in ulcerative colitis. Lancet. 1998;351(9115):1555.
112. Langmead L, Feakins R, Goldthorpe S, et al. Randomized, double blind, placebo-controlled trial of oral aloe vera gel for active ulcerative colitis. Aliment Pharmacol Ther. 2004;19:739–747.
113. Krebs S, Omer B, Omer N. Wormwood (Artemisia absinthium) suppresses tumor necrosis factor alpha and accelerates healing in patients with Crohn’s disease – A controlled clinical trial. Phytomedicine. 2010;17:305–309.
Biliary system
Apart from their use to provide non-specific support for recuperation and repair, specific phytotherapeutic strategies include the following.
Because of the use of secondary plant products, particular caution is necessary in applying phytotherapy to:
Whereas cholesterol accounts for more than 90% to 95% of the sterols in bile, bile acids and their salts are the most important solutes; they are essential in the management of cholesterol levels and themselves help determine the extent of bile flow. Bile acids are synthesised from cholesterol by the liver. There are three groups. Primary bile acids, in humans mainly cholic and chenodeoxycholic acids and their salts, are produced directly. Secondary bile salts are created by the action of intestinal bacteria on primary bile salts with deoxycholate and lithocholate being formed from cholate and chenodeoxycholate, respectively. Tertiary bile salts are the result of modification of secondary bile salts by intestinal flora or hepatocytes; in humans these include the sulphate ester of lithocholate and ursodeoxycholate and the 7-beta-epimer of chenodeoxycholate.1
Bile flow rates and composition are subject to a wide variety of neural, endocrine and paracrine influences. One of the main stimulants of bile flow are bile acids themselves, either in their primary form or reabsorbed as secondary or tertiary forms in the enterohepatic circulation (see below). The cholagogue effects of bile acids have led to their prescription in hepatobiliary disorders. One derivative, ursodeoxycholic acid, has been shown in controlled clinical studies to be a useful agent in the management of patients with primary biliary cirrhosis, autoimmune chronic active hepatitis2 and cystic fibrosis.3
Infective conditions may lead to cholestasis or reduced bile flow with symptoms including jaundice, pruritus and steatorrhoea. Chronic alcoholics may have hypotonic gallbladder, with increased speed of bile secretion and low biliary levels of cholic acid, cholesterol and bilirubin. Patterns of bile stagnation can occur with increasing severity of alcoholism, especially when associated with cirrhosis.4 This effect has been attributed to the effects of lipopolysaccharide endotoxins.5
The role of inflammatory bowel disease in inducing cholestasis is also well established.6 In one study serum cholestanol/cholesterol proportions were determined in 79 patients with inflammatory bowel (colonic and ileal) diseases, such as ulcerative colitis and Crohn’s disease, and 23 with irritable bowel syndrome as controls. The findings suggested that the increased cholestanol proportion in colonic inflammatory bowel diseases is determined mainly by impaired biliary elimination of this sterol, while in ileal disease the dominating change in sterol balance is activated cholesterol synthesis. Increased serum cholestanol is a novel finding in colonic inflammatory bowel diseases, apparently indicating the presence of subclinical cholestasis in a marked number (20% to 50%) of inflammatory bowel disease patients.7
Therapeutic stimulation of bile flow could thus be justified in the management and treatment of any of the above circumstances (see below).
It appears that a substantial amount of urate is also eliminated by the biliary route in humans. Gout and other urate-associated conditions linked to decreased renal urate excretion may therefore benefit from measures that increase biliary urate excretion.8
Because it is known that high concentrations of bile acids are cytotoxic, it has been speculated that their raised presence in serum and tissues in hepatobiliary diseases contributes to the pathological progression of these disorders. Bile acids are a causative factor in chronic gastritis.9 Evidence is that oral administration of ursodeoxycholate, being a relatively non-toxic bile acid, can replace more hydrophobic hepatotoxic bile acids in the circulating pool and, by doing so, ameliorate the harmful effects of the latter.10
An important aspect of bile acid function and toxicity is the constant reabsorption from the intestine into the portal circulation and back to the liver and biliary system – the enterohepatic circulation. Both primary and secondary bile acids are involved in this recycling. In modern medicine a high degree of recycling is assumed. Only 1% of bile acids are lost in the faeces and it was calculated that bile acids are recirculated about 12 times per day.11 However, it is likely that humans living a more primitive lifestyle with much higher levels of fibre intake had lower reabsorption rates. The implications of enterohepatic circulation are best understood with reference to the kinetics of drugs such as the morphine alkaloids and digoxin, which are largely eliminated from the body through the bile. Increased retention of bile in the enterohepatic circulation is known to increase the half-life of these and other drugs in the body. It is likely, therefore, that the level of bile products (with the formation of tertiary bile acids) and other potentially toxic metabolites may increase, unless the enterohepatic circulation is as low as possible. In practice, this is most likely to follow a relatively fast intestinal transit time, associated with a high-fibre diet.
Although they are generally cholagogic, the presence of bile acids in the enterohepatic cycle acts to decrease the hepatic production of cholesterol, presumably through a process of negative feedback, and this is likely to control excessive cholesterol secretion in gallstone conditions. As with other hepatic conditions, replacement therapy with bile acids such as chenodeoxycholic and ursodeoxycholic acids is promoted as a treatment, leading even to dissolution of existing gallstones.12
Of wider significance is the finding that reduction in the absorption of bile acids from the gut, associated with, for example, diarrhoea, leads to an increase in cholesterol synthesis and cholesterol esterification rate by the liver.13,14
Biliary cholesterol excretion is directly linked to two major pathological issues, namely atherosclerotic cardiovascular disease (ACVD) and cholesterol gallstones. In ACVD biliary cholesterol secretion is the final step in the reverse cholesterol transport (RCT) pathway, the transport of peripheral cholesterol back to the liver for excretion.15 For RCT, enhanced biliary secretion of cholesterol is desirable, although this can lead to biliary cholesterol supersaturation and gallstones. The most relevant source of cholesterol secreted into bile is cholesterol derived from plasma lipoproteins, with HDL (high density lipoprotein) the preferential contributor.15 However, definitive studies exploring the underlying metabolic pathways are still lacking, although ABC (ATP binding cassette) transporters are known to be involved: specifically ABCB11 for bile acid transport into bile, ABCB4 for phospholipids and ABCG5/G8 for cholesterol.15
Both primary and secondary bile acids have secretagogue effects on the intestinal mucosa, changing net fluid transport across the villi from absorption to secretion.16 There is also an atropine-inhibited (i.e. cholinergic) stimulation of intestinal contractions17 and an increased mucosal vasodilation (blood flow increasing by around 50%), which is not inhibited by atropine.18 Secondary bile acids in particular are thought to increase permeability at the zonulae occludentes that binds endothelial cells together at their luminal borders, so that normal subepithelial hydrostatic pressure is raised sufficiently to reverse net sodium, chloride and water absorption to net secretion.19
Bile has sometimes been referred to as the body’s own laxative or ‘endolaxative’.20 Indeed, it is established that bile acids (especially secondary bile acids – see below) can be responsible for bowel looseness and their effects should be borne in mind in cases of unexplained chronic diarrhoea.21,22
Bile has a wider range of actions on the intestinal mucosa. Where there is clear reduction in bile levels, there is a reduction in thickness of the mucus blanket, reduced numbers of mucus-associated enterocytes (suggesting a reduced endothelial turnover rate) and lymphocytes and increased populations of bacterial organisms. The implication is that normal bile function is a vital part of the body’s gut-related defences.23 It has also been demonstrated that bacterial endotoxin absorption is increased in the absence of bile salts from the intestine.24
The generally positive functions of primary bile acids become rather more mixed in their effects once bacterial deconjugation and dehydroxylation occur. Secondary bile acids are produced at a very early age – the process is clearly under way even in month-old infants25 – and are an entirely normal range of metabolites. It is clear that most bacterial deconjugation occurs in the colon26 but in various less ideal circumstances invasive bacterial populations can lead to secondary bile product formation in the small intestine.
Secondary bile acids have a decidedly irritating effect on the intestinal wall. Exposure of the intestinal wall to deconjugated bile acids stimulates local inflammatory mechanisms, accompanied by the release of prostaglandin E2 and leukotriene C4. Such effects are particularly pronounced if there is already latent or active inflammatory disease of the intestinal wall, particularly in small intestinal Crohn’s disease.27 The irritant effect of secondary bile products is especially apparent if their quantities are increased due to stasis in the small intestine. Among a number of ultrastructural alterations to the intestinal mucosa, they increase the numbers of lysosomal vascular structures, fused microvilli and dilated endoplasmic reticulum, which among other implications leads to a reduced absorption of solutes including glucose and other carbohydrates28,29 and, significantly, fluid absorption:30 diarrhoea is thus possible. Secondary bile metabolites substantially increase the absorption rates of urea and oxalates from the gut.31
There are more insidious potential effects too. Secondary bile acids and their metabolites increase colonic cell proliferation.32 The carcinogenic effect is clearly linked to changes in the nature of bacterial populations in the gut, rather than to the nature of the bile acids or indeed other starting materials in the gut.33 However, there is also little doubt that decreased reabsorption of bile acids (for example, as seen with increasing old age) does increase the likelihood that carcinogenic and other pathogenic bile metabolites will be produced.34
Dietary factors are known to affect the balance between intestinal flora and bile metabolism. For example, consumption of sugars was shown in nine volunteers on a crossover basis to significantly prolong transit time through the colon and significantly raise the faecal levels of both primary and secondary bile metabolites.35 On the other hand, the consumption of 16 g of wheat bran a day on a double blind, 6-month crossover basis by ulcerative colitis sufferers in remission was shown to decrease the faecal concentration of bile acids by almost half. No such effect was observed with psyllium seed.36
There are some potential benefits in bacterial action on bile acids. Anaerobic bacteria, for example, can produce volatile fatty acids known to non-specifically inhibit pathogenic bacterial populations.37
Disturbed and pathological states can change the dynamics of bile and other intestinal relationships. In malnutrition, for example, morphological changes in the intestinal wall lead to increased sensitivity to the effects of secondary bile acids, poor absorption of fats and other nutrients, all of which is compounded by changes in intestinal flora.38
The impact of inflammatory intestinal diseases like Crohn’s is even more pronounced. The damage induced by the disease on the intestinal wall leads to reduced bile acid reabsorption and compensatory increased cholesterol synthesis by the liver,39 a reaction that probably explains the high level of biliary disease such as gallstones in sufferers from Crohn’s.40,41 The link between inflammatory bowel disease and cholestasis has already been mentioned, and Crohn’s disease in particular is linked with disturbed bile metabolism and gallstones.42 Similar negative impact on enterohepatic circulation follows small intestinal resection, which has been shown to lead to increased synthesis of both bile acids and cholesterol.43
The association between biliary and intestinal functions is further highlighted in the condition primary sclerosing cholangitis, a disease characterised by inflammation and obliterative fibrosis of bile ducts. In 70% of cases it is associated with ulcerative colitis. In about two-thirds, there are circulating IgG antibodies to a peptide shared by epithelial cell walls in both bile ducts and colon. Another suggested cause is portal bacteraemia secondary to a diseased bowel wall. The addition of bile acids to the gut has been proposed as a treatment.44–46
A useful insight has been made in studies of the metabolism of plant sterols. Plant sterols are structurally similar to cholesterol but, because of poor intestinal absorption, are ordinarily not present in the liver. However, there does appear to be competition in the movement of plant sterols and cholesterol. For example, high plant sterol consumption appears to lower blood cholesterol levels,47 especially in the short term,48 and the proportions of plant sterols are significantly lower in cholesterol-rich gallstones than in bile (and stones with low cholesterol content are proportionately richer in plant sterols).49 One sterol studied, sitostanol, parallels the secretion from and distribution of cholesterol in the liver (for example, both requiring bile salts for secretion in bile) so that it can be used as a physiologic analogue of unesterified cholesterol to trace the transport of sterols through the liver.50 Such studies, for example, indicate that HDLs are necessary along with bile acids for cholesterol elimination in bile.51 The use of plant sterols (in this case campesterol and sitosterol) as markers of cholesterol absorption and biliary secretion was also seen in a study referred to above, showing subclinical cholestasis as a feature of inflammatory bowel disease.52
When serum concentrations and metabolism of cholesterol were studied in human vegetarians, cholesterol absorption was found to be normal and synthesis was slightly enhanced, though without increase in serum cholesterol precursors. The serum concentrations of total and low-density lipoprotein (LDL)–cholesterol were decreased but, in addition to the obvious lower intake of cholesterol itself, it appeared that the higher intake of plant sterols interfered with cholesterol absorption and thus increased endogenous cholesterol synthesis. Thus, cholesterol saturation and bile acid composition of the bile were not changed. Biliary excretion of plant sterols was apparently relatively inefficient.53
Interactions between cholesterol transport and plant constituents extend to another major group. Saponins have been implicated in interference with the absorption of cholesterol, bile acids and fats, leading to reduced animal growth, but also have shown potential in the reduction of blood cholesterol levels.54 There is evidence of interference with the absorption of vitamins A and E.55 Their cholesterol-lowering effect may also be linked to their binding of bile salts and increasing their faecal excretion, thus increasing bile salt synthesis from endogenous cholesterol.56 Further studies to investigate the effects of saponins on bile, cholesterol and lipid metabolism are clearly warranted (see also Chapter 2). One steroidal sapogenin that has been studied, diosgenin, is, like the sterols, also similar enough in structure to cholesterol to interfere with its esterification in the liver.57 This may contribute to the marked increase observed in biliary cholesterol relative to phospholipids when it was fed for 7 days to rats (see Chapter 2).58
There is a traditional differentiation made between cholagogues and choleretics. The former are agents that stimulate the release of bile that has already been formed in the biliary system. Bile acids are the main endogenous cholagogues and fatty foods the most obvious exogenous factors.
Choleretics stimulate bile production by hepatocytes and some have effective cholagogue properties as well. Cholecystokinin, secretin and some of the other humoral agents are involved endogenously. Bitters and some of the botanical agents referred to below are likely to have choleretic activity (see Chapter 2).
There is very little interest in choleretic and cholagogue treatments in conventional medicine, at least in the English-speaking world. Among agents incidentally discovered, NSAIDs, especially aspirin (in one study at a level of 100 mg/kg), cause choleresis in animals.59 Magnesium sulphate (Epsom salts), sometimes used for constipation, has at doses of 500 mg been shown to exert a direct effect on the motor activity of the gallbladder in dogs.60
Research on herbal choleretics and cholagogues has largely come from Germany and Eastern Europe. It is not comprehensive and most practice in this area is informed by traditional reputation. Given the difficulty in knowing what actually happens in the liver and the potential risks of counterproductive treatments (see below), this is not an ideal situation.
Among the work that has been done, it has been shown that the ethanolic extract of Chelidonium majus (greater celandine) in isolated liver culture significantly caused choloresis by increasing bile acid-independent flow,61 and there is indication of activity in this area in clinical trials (see monograph). A survey of the literature shows that Cynara scolymus (artichoke) possesses choleretic, diuretic and hypocholesterolaemic properties (see monograph). The main active components of this plant are mono- and dicaffeoylquinic acids, flavonoids and sesquiterpenes. The most suitable raw material is fresh leaves in the plant’s first year of growth.62 Recent focus has been on the contribution of the claimed choleretic activity to cholesterol reduction. A Cochrane review63 of three good-quality double blind studies points to a modest effect on total and LDL-cholesterol levels, although not at sufficient levels to recommend to prescribers. In one of the more significant placebo-controlled studies, involving 75 people in the UK, total cholesterol was reduced by 42% in the group receiving 1280 mg of standardised artichoke leaf extract per day for 12 weeks, whereas the levels in the placebo group actually rose.64 The choleretic activity of globe artichoke leaf has also been confirmed in clinical studies (see monograph). Phenolic acids in Mentha longifolia were found to possess significant in vivo choleretic and CNS stimulating effects65 and peppermint leaf is traditionally regarded as a cholagogue. Turmeric (Curcuma longa) is also a clinically relevant cholagogue and choleretic herb (see monograph).
• Berberis vulgaris (barberry), Berberis aquifolium (Oregon grape), Chelidonium (greater celandine), Chelone (balmony), Chionanthus (fringe-tree), Euonymus atropurpureus (wahoo), Taraxacum (dandelion), Veronicastrum (black root), Peumus (boldo), Curcuma longa (turmeric) and Cynara (globe artichoke).
The effects of choleretic and cholagogue agents may be different in the diseased liver than the response produced in the normal liver. For example, experimental evidence suggests that the use of choleretic agents where hepatobiliary damage (e.g. cholangitis) is caused by obstructive jaundice might further depress hepatic functions.66
The use of choleretics and cholagogues is either contraindicated or at least inappropriate in the following:
• Obstructed bile ducts (due to impacted gallstones, cholangitis or cancer of the bile duct or pancreas)
• Unconjugated hyperbilirubinaemia (jaundice following haemolytic diseases, hereditary disease such as Gilbert’s and Crigler-Najjar syndromes)
• Acute or severe hepatocellular disease (for example, following viral hepatitis, cirrhosis, adverse reactions to drugs, such as anaesthetics, steroids, oestrogen, chlorpromazine)
• Septic cholecystitis (where there is a risk of peritonitis)
• Hepatic cancer (although hepatoprotective herbs that also have some choleretic activity, such as Silybum marianum, can be appropriate, especially for secondary tumours on the liver).
Stimulating bile flow has been seen as one of the main eliminative strategies in traditional medicine, reflecting the importance attached by even the most primitive cultures to the role of the liver (the name of the organ was often as evocative as it is in English).
However, bile stimulation was often accompanied by vigorous approaches to eliminating it from the gut as well; the almost universal reliance in folk medicine on emetics and purgatives as first-resort approaches to the treatment of acute disease almost certainly had the effect, intended or otherwise (and in the case of emetics it was often intended), of radically removing bile from the body. It is now easy in modern medicine to dismiss the use of such drastic procedures as useless or dangerous but, unlike the use of bleeding, which was often likely to be counterproductive, emesis and catharsis were almost prehuman measures and established themselves over millennia in the most demanding court of efficacy, the survival from acute disease.
Given what is now known of the retentive qualities of the enterohepatic cycle, the certainty that modern diets and lifestyle have significantly lengthened intestinal transit time compared to that of early humans, thus extending enterohepatic recycling further, it is likely that many modern practitioners might look with some envy at their forebears’ ability to get rid of this pool of potentially or actually toxic metabolites. However, emesis and catharsis were seen as an option for the most robust constitutions and, apart from the obvious inappropriateness, they are contraindicated in the more chronic and low-vitality conditions most often seen in the modern clinic.
Modern techniques to eliminate the bile pool generally involve dietary and other measures to decrease intestinal transit time combined with the use of choleretics and cholagogues. Some of the latter actually have laxative effects in any case, either because they contain the appropriate constituents or, more often, because the release of more bile is in itself laxative.
Choleretics and cholagogues are best taken before meals, preferably about 30 minutes, but immediately before will suffice. As many rely at least in part on the effect of bitter constituents, they are best taken in fluid form.
Choleretics and cholagogues may also be usefully applied in some cases (depending on other factors) of:
• chronic constipation (not due to intestinal spasm nor responding to conventional measures)
• dysbiotic conditions of the gut
• autoimmune diseases (especially where associated with any of the above – see relevant sections, especially in Chapter 8).
Gallstones are formed when cholesterol and other solute levels in bile reach supersaturated concentrations and when the normal glassy-smooth surfaces of the gallbladder are compromised, often by infection. Bile is often supersaturated after a night’s metabolism and before breakfast: this could explain the naturopathic practice of recommending lemon juice (a liver and gallbladder stimulant) before breakfast. The concentration of bile also appears to be related to intestinal activity and slow intestinal transit has been linked to gallstone formation in normal-weight women67 and in other studies.68
Certain risk factors for gallstones are inherent: being female, increasing age and ethnicity/family history (genetic traits). Others are modifiable: obesity, metabolic syndrome, hypertriglyceridaemia, rapid weight loss, diet (low in fibre and high in refined carbohydrates and saturated fat) and certain diseases (cirrhosis and Crohn’s disease).69
Findings largely from in vitro and in vivo studies suggest that infection, inflammation and the response of the immune system can also influence the pathogenesis of cholesterol gallstones.70 Supersaturated bile can lead to biliary sludge, composed of agglomerated cholesterol crystals. However, human studies have shown that biliary sludge does not necessarily lead to gallstone formation.70 The missing link could be infection and/or inflammation that seeds stone formation.
Most patients with gallstones have no symptoms and current medical thinking is that there is no distinct advantage in treating asymptomatic gallstones.71 Small stones are more dangerous than large as they can cause pancreatitis.72 Oral dissolution therapy with bile salts is still used as a treatment, but is reserved for patients with non-calcified cholesterol gallstones, a patent cystic duct and for those who do not require urgent surgery.71 These considerations also define the conditions for successful herbal treatment of gallstones. Patients who receive conventional oral therapy usually have a high rate of gallstone recurrence. This underlines the need in herbal therapy for long-term treatment concurrent with appropriate dietary and lifestyle changes.
A key outcome of phytotherapy for cholesterol gallstones is that it can render symptomatic gallstones quiescent. However, its greatest value here will be for stones that are not calcified in a functional gallbladder.
The essential elements of treatment are as follows:
• Bitter herbs to improve digestive and gallbladder function, such as Artemisia absinthium (wormwood) or Gentiana lutea (gentian)
• Choleretic herbs to improve bile flow, such Chelidonium (greater celandine), Cynara (globe artichoke), Taraxacum radix (dandelion root) and Silybum (St Mary’s thistle)
• Cholagogue herbs to improve gallbladder motility, such as Chelidonium (greater celandine), Cynara and Mentha piperita (peppermint). A proprietary terpene mixture similar to essential oil of peppermint has been shown to dissolve gallstones73
• Spasmolytic herbs, selected from Viburnum opulus (cramp bark), Corydalis ambigua, Matricaria (chamomile) and Mentha piperita, can help to relieve gallbladder pain
• Long-term use of herbs containing steroidal saponins such as Dioscorea (wild yam) and Smilax (sarsaparilla) is best avoided, since these herbs may increase cholesterol levels in bile.74
A short course of copious olive oil and lemon juice is often recommended to discharge gallstones, although this is controversial.75,76 However, such a therapy should only be attempted if the gallstones are not calcified, the cystic duct is patent, the gallbladder is functional and herbal therapy to soften the stones has been given for at least 6 months.
The above herbal approach can also be used for functional gallbladder disorder (gallbladder dyskinesia), which is the recurrence of abdominal pain resembling gallbladder pain in the absence of gallstones.77
Case history
A male patient aged 69 had been experiencing recurrent attacks of biliary pain for several months. A blood test showed the presence of high levels of bilirubin, perhaps due to temporary obstruction caused by the passage of a stone, and tests revealed gallbladder inflammation and gallstones. The patient was offered surgery but wanted to try herbal treatment first.
He was advised to follow a low fat diet and the following formula was prescribed:
Silybum marianum | 1:1 | 25 mL |
Cynara scolymus | 1:2 | 25 mL |
Taraxacum officinale radix | 1:2 | 20 mL |
Picrorrhiza kurroa | 1:2 | 10 mL |
Mentha piperita | 1:2 | 20 mL |
total | 100 mL |
Dose: 5 mL with water three times a day.
After a few months of treatment all symptoms had abated. The patient continued treatment for another 6 months, during which time he was free of symptoms. Since that time (several years) he has not had any herbal treatment but still remains free of gallbladder symptoms.
Case history
An overweight female patient aged 61 years developed recurrent symptoms of nausea, vomiting and upper abdominal pain and was diagnosed as having gallstones. A low fat diet was recommended with avoidance of fried food.
The following herbal formula was prescribed and rapidly ameliorated her symptoms to the point it was no longer required after 6 weeks of use:
Taraxacum officinale radix | 1:2 | 20 mL |
Silybum marianum | 1:1 | 20 mL |
Cynara scolymus | 1:2 | 25 mL |
Matricaria recutita | 1:2 | 20 mL |
Corydalis ambigua | 1:2 | 20 mL |
105 mL |
Dose: 8 mL with water three times a day initially until symptoms subsided, and then twice a day thereafter.
References
1. Hay DW, Carey MC. Chemical species of lipids in bile. Hepatology. 1990;12(3 pt 2):6–14.
2. Heathcote EJ, Cauch-Dudek K, Walker V, et al. The Canadian multicenter double blind randomized controlled trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology. 1994;19(5):1149–1156.
3. Van Demeeberg PC, Houwen RHJ, Sinaasappel M, et al. Low-dose versus high-dose in cystic fibrosis-related cholestatic liver disease: results of a randomized study with 1-year follow-up. Scand J Gastroenterol. 1997;32(4):369–373.
4. Mikhailovskaya AY, Loranskaya TI, Vasilevskaya LS. Liver bile-secreting function in chronic alcoholics and means of correction of its disorders. Voprosy Pitaniya. 1995;5:34–36.
5. Trauner M, Nathanson MH, Rydberg SA, et al. Endotoxin impairs biliary glutathione and HCO-3-excretion and blocks the choleretic effect of nitric oxide in rat liver. Hepatology. 1997;25(5):1184–1191.
6. Huang CS, Lichtenstein DR. Treatment of biliary problems in inflammatory bowel disease. Curr Treat Options Gastroenterol. 2005;8(2):117–126.
7. Hakala K, Vuoristo M, Miettinen TA. Serum cholestanol, cholesterol precursors and plant sterols in different inflammatory bowel diseases. Digestion. 1996;57(2):83–89.
8. Kountouras J, Magoula I, Tsapas G, Liatsis I. The effect of mannitol and secretin on the biliary transport of urate in humans. Hepatology. 1996;23(2):229–233.
9. Kolarski V, Petrova-Shopova K, Vasileva E, et al. Erosive gastritis and gastroduodenitis – clinical, diagnostic and therapeutic studies. Vutr Boles. 1987;26(3):56–59.
10. Maillette de Buy Wenniger L, Beuers U. Bile salts and cholestasis. Dig Liver Dis. 2010;42(6):409–418.
11. Lester R, Zimniak P. True transport: one or more sodium-dependent bile acid transporters? Hepatology. 1993;18(5):1279–1282.
12. Hofmann AF. Bile acids as drugs: principles, mechanisms of action and formulations. Ital J Gastroenterol. 1995;27(2):106–113.
13. Akerlund JE, Reihner E, Angelin B, et al. Hepatic metabolism of cholesterol in Crohn’s disease. Effect of partial resection of ileum. Gastroenterology. 1991;100(4):1046–1053.
14. Stahlberg D, Reihner E, Angelin B, Einarsson K. Interruption of the enterohepatic circulation of bile acids stimulates the esterification rate of cholesterol in human liver. J Lipid Res. 1991;32(9):1409–1415.
15. Dikkers A, Tietge UJ. Biliary cholesterol secretion: more than a simple ABC. World J Gastroenterol. 2010;16(47):5936–5945.
16. Gaginella TS, Haddad AC, Go VL, Phillips SF. Cytotoxicity of ricinoleic acid (castor oil) and other intestinal secretagogues on isolated intestinal epithelial cells. J Pharmacol Exp Ther. 1977;201(1):259–266.
17. Karlstrom L. Evidence of involvement of the enteric nervous system in the effects of sodium deoxycholate on small-intestinal transepithelial fluid transport and motility. Scand J Gastroenterol. 1986;21(3):321–330.
18. Karlstrom L. Mechanisms in bile salt-induced secretion in the small intestine. An experimental study in rats and cats. Acta Physiol Scand. 1986;549:1–48.
19. Wanitschke R. Intestinal filtration as a consequence of increased mucosal hydraulic permeability. A new concept for laxative action. Klin Wochenschr. 1980;58(6):267–278.
20. Abrahamsson H, Ostlund-Lindqvist AM, Nilsson R, et al. Altered bile acid metabolism in patients with constipation-predominant irritable bowel syndrome and functional constipation. Scand J Gastroenterol. 2008;43(12):1483–1488.
21. Kurien M, Evans KE, Leeds JS, et al. Bile acid malabsorption: an under-investigated differential diagnosis in patients presenting with diarrhea predominant irritable bowel syndrome type symptoms. Scand J Gastroenterol. 2011;46(7–8):818–822.
22. Ford GA, Preece JD, Davies IH, Wilkinson SP. Use of the SeHCAT test in the investigation of diarrhoea. Postgrad Med J. 1992;68(798):272–276.
23. Kalambaheti T, Cooper GN, Jackson GD. Role of bile in non-specific defence mechanisms of the gut. Gut. 1994;35(8):1047–1052.
24. Cahill CJ, Pain JA, Bailey ME. Bile salts, endotoxin and renal function in obstructive jaundice. Surg Gynecol Obstet. 1987;165(6):519–522.
25. Jonsson G, Midtvedt AC, Norman A, Midtvedt T. Intestinal microbial bile acid transformation in healthy infants. J Pediatr Gastroenterol Nutr. 1995;20(4):394–402.
26. Bruwer M, Stern J, Stiehl A, Herfarth C. Changes in fecal bile acid excretion after proctocolectomy. Z Gastroenterol. 1996;34(2):105–110.
27. Casellas F, Guarner F, Antolin M, et al. Abnormal leukotriene C4 released by unaffected jejunal mucosa in patients with inactive Crohn’s disease. Gut. 1994;35(4):517–522.
28. Wehman HJ, Lifshitz F, Teichberg S. Effects of enteric microbial overgrowth on small intestinal ultrastructure in the rat. Am J Gastroenterol. 1978;70(3):249–258.
29. Lifshitz F, Wapnir RA, Wehman HJ, et al. The effects of small intestinal colonization by fecal and colonic bacteria on intestinal function in rats. J Nutr. 1978;108(12):1913–1923.
30. Fukushima T, Ishiguro N, Tsujinaka Y, et al. Bile acid deconjugation in intestinal obstruction studied by breath test. Jpn J Surg. 1977;7(2):73–81.
31. Emmett M, Guirl MJ, Santa Ana CA, et al. Conjugated bile acid replacement therapy reduces urinary oxalate excretion in short bowel syndrome. Am J Kidney Dis. 2003;41(1):230–237.
32. Parsonnet J. Bacterial infection as a cause of cancer. Environ Health Perspect. 1995;103(suppl 8):263–268.
33. Kanazawa K, Konishi F, Mitsuoka T, et al. Factors influencing the development of sigmoid colon cancer. Bacteriologic and biochemical studies. Cancer. 1996;77(8 suppl):1701–1706.
34. Salemans JM, Nagengast FM, Tangerman A, et al. Effect of ageing on postprandial conjugated and unconjugated serum bile acid levels in healthy subjects. Eur J Clin Invest. 1993;23(3):192–198.
35. Kruis W, Forstmaier G, Scheurlen C, Stellaard F. Effect of diets low and high in refined sugars on gut transit, bile acid metabolism, and bacterial fermentation. Gut. 1991;32(4):367–371.
36. Ejderhamn J, Hedenborg G, Strandvik B. Long-term double blind study on the influence of dietary fibres on faecal bile acid excretion in juvenile ulcerative colitis. Scand J Clin Lab Invest. 1992;52(7):697–706.
37. Tazume S, Ozawa A, Yamamoto T, et al. Ecological study on the intestinal bacteria flora of patients with diarrhea. Clin Infect Dis. 1993;16(suppl 2):77–82.
38. Behar M. The role of feeding and nutrition in the pathogeny and prevention of diarrheic processes. Bull Pan Am Health Organ. 1975;9(1):1–9.
39. Ejderhamn J, Rafter JJ, Strandvik B. Faecal bile acid excretion in children with inflammatory bowel disease. Gut. 1991;32(11):1346–1351.
40. Hutchinson R, Tyrrell PN, Kumar D, et al. Pathogenesis of gall stones in Crohn’s disease: an alternative explanation. Gut. 1994;35(1):94–97.
41. Murray FE, McNicholas M, Stack W, O’Donoghue DP. Impaired fatty-meal-stimulated gallbladder contractility in patients with Crohn’s disease. Clin Sci (Colch). 1992;6:689–693.
42. Lapidus A, Akerlund JE, Einarsson C. Gallbladder bile composition in patients with Crohn’s disease. World J Gastroenterol. 2006;12(1):70–74.
43. Akerlund JE, Bjorkhem I, Angelin B, et al. Apparent selective bile acid malabsorption as a consequence of ileal exclusion: effects on bile acid, cholesterol, and lipoprotein metabolism. Gut. 1994;35(8):1116–1120.
44. Stiehl A. Ursodeoxycholic acid in the treatment of primary sclerosing cholangitis. Ann Med. 1994;26(5):345–349.
45. Boberg KM, Lundin KE, Schrumpf E. Etiology and pathogenesis in primary sclerosing cholangitis. Scand J Gastroenterol. 1994;204:47–58.
46. Mandal A, Dasgupta A, Jeffers L, et al. Autoantibodies in sclerosing cholangitis against a shared peptide in biliary and colon epithelium. Gastroenterology. 1994;106(1):185–192.
47. Deng R. Food and food supplements with hypocholesterolemic effects. Recent Pat Food Nutr Agric. 2009;1(1):15–24.
48. Gupta AK, Savopoulos CG, Ahuja J, et al. Role of phytosterols in lipid-lowering: current perspectives. QJM. 2011;104(4):301–308.
49. Miettinen TE, Kesaniemi YA, Gylling H, et al. Noncholesterol sterols in bile and stones of patients with cholesterol and pigment stones. Hepatology. 1996;23(2):274–280.
50. Robins SJ, Fasulo JM, Pritzker CR, Patton GM. Hepatic transport and secretion of unesterified cholesterol in the rat is traced by the plant sterol, sitostanol. J Lipid Res. 1996;37(1):15–21.
51. Robins SJ, Fasulo JM. High density lipoproteins, but not other lipoproteins, provide a vehicle for sterol transport to bile. J Clin Invest. 1997;99(3):380–384.
52. Hutchinson R, Tyrrell PN, Kumar D, et al. Pathogenesis of gall stones in Crohn’s disease: an alternative explanation. Gut. 1994;35(1):94–97.
53. Vuoristo M, Miettinen TA. Absorption, metabolism, and serum concentrations of cholesterol in vegetarians: effects of cholesterol feeding. Am J Clin Nutr. 1994;59(6):1325–1331.
54. Francis G, Kerem Z, Makkar HPS, Becker K. The biological action of saponins in animal systems: a review. Br J Nutr. 2002;88:587–605.
55. Jenkins KJ, Atwal AS. Effects of dietary saponins on fecal bile acids and neutral sterols, and availability of vitamins A and E in the chick. J Nutr Biochem. 1994;5(3):134–137.
56. Oakenfull D, Sidhu GS. Could saponins be a useful treatment for hypercholesterolaemia? Eur J Clin Nutr. 1990;44:79–88.
57. Son IS, Kim JH, Sohn HY, et al. Antioxidative and hypolipidemic effects of diosgenin, a steroidal saponin of yam (Dioscorea spp.), on high-cholesterol fed rats. Biosci Biotechnol Biochem. 2007;71(12):3063–3071.
58. Roman ID, Thewles A, Coleman R. Fractionation of livers following diosgenin treatment to elevate biliary cholesterol. Biochim Biophys Acta. 1995;1255(1):77–81.
59. Nussinovitch M, Zahavi I, Marcus H, et al. The choleretic effect of nonsteroidal anti-inflammatory drugs in total parenteral nutrition-associated cholestasis. Isr J Med Sci. 1996;32(12):1262–1264.
60. Sterczer A, Voros K, Karsai F. Effect of cholagogues on the volume of the gallbladder of dogs. Res Vet Sci. 1996;60(1):44–47.
61. Táborská E, Bochoráková H, Dostál J, Paulová H. The greater celandine (Chelidonium majus L.) – review of present knowledge. Ceska Slov Farm. 1995;44(2):71–75.
62. Dranik LI, Dolganenko LG, Slapke J, Thoma H. Chemical composition and medical usage of Cynara scolymus L. Rastitel’nye Resursy. 1996;32(4):98–104.
63. Wider B, Pittler MH, Thompson-Coon J, Ernst E. Artichoke leaf extract for treating hypercholesterolemia. Cochrane Database Syst Rev. 2009;4:CD000335.
64. Bundy R, Walker AF, Middleton RW, et al. Artichoke leaf extract (Cynara scolymus) reduces plasma cholesterol in otherwise healthy hypercholesterolemic adults: a randomized, double blind placebo controlled trial. Phytomedicine. 2008;15:668–675.
65. Mimica-Dukic N, Jakovljevic V, Mira P, et al. Pharmacological study of Mentha longifolia phenolic extracts. Int J Pharmacog. 1996;34(5):359–364.
66. Ishibashi H, Komori A, Shimoda S, et al. Risk factors and prediction of long-term outcome in primary biliary cirrhosis. Intern Med. 2011;50(1):1–10.
67. Heaton KW, Emmett PM, Symes LJ, et al. An explanation for gallstones in normal-weight women: slow intestinal transit. Lancet. 1993;341(8836):8–10.
68. Venneman NG, van Erpecum KJ. Pathogenesis of gallstones. Gastroenterol Clin North Am. 2010;39(2):171–183.
69. Stinton LM, Myers RP, Shaffer EA. Epidemiology of gallstones. Gastroenterol Clin North Am. 2010;39(2):157–169.
70. Maurer KJ, Carey MC, Fox JG. Roles of infection, inflammation, and the immune system in cholesterol gallstone formation. Gastroenterology. 2009;136(2):425–440.
71. Portincasa P, Di Ciaula A, Wang HH, et al. Medicinal treatments of cholesterol gallstones: old, current and new perspectives. Curr Med Chem. 2009;16(12):1531–1542.
72. Sanders G, Kingsnorth AN. Gallstones. BMJ. 2007;335(7614):295–299.
73. Bell GD, Doran J. Gall stone dissolution in man using an essential oil preparation. BMJ. 1(6155), 1979. 24
74. Thewles A, Parslow RA, Coleman R. Effect of diosgenin on biliary cholesterol transport in the rat. Biochem J. 1993;291(3):793–798.
75. Savage AP, O’Brien T, Lamont PM. Adjuvant herbal treatment for gallstones. Br J Surg. 1992;79(2):168.
76. Sies CW, Brooker J. Could these be gallstones? Lancet. 2005;365(9468):1388.
77. Hansel SL, DiBaise JK. Functional gallbladder disorder: gallbladder dyskinesia. Gastroenterol Clin North Am. 2010;39(2):369–379. x
The liver
Apart from their use to provide non-specific support for recuperation and repair, specific phytotherapeutic strategies include the following.
• postpartum symptoms of liver distress, such as after consumption of fatty foods and alcohol
Because of its use of secondary plant products, particular caution is necessary in applying phytotherapy to primary and secondary liver carcinoma.
Given its relative lack of attention in most medical books, the liver has an enormous hidden importance. The hub of many of the body’s biochemical pathways, and first processor of dietary metabolites, it also has established roles in modulating the immune and endocrine systems (any hormone has two influences on its blood levels, the rate of production and the rate of breakdown in the liver – this is an often varied factor1), and cholesterol and blood sugar levels. With its associate Kupffer cells it is a vital defence wall in protecting the body against immunological threat from foodstuffs. In its biliary production it has a key influence on digestion itself and bowel flora and function. All these central roles support the primitive appreciation for the liver as a centre-piece in traditional systems of medicine, and even its name in many languages.
Most obvious is the liver’s primary role in detoxification, both of ingested molecules (xenobiotics) and internal metabolites and agents such as hormones. There are two phases of hepatic biotransformation involved. During phase I, enzymatic-induced oxidation, reduction or hydrolysis generates a reactive site on the substrate molecule; in phase II a water-soluble (hydrophilic) group is conjugated with this reactive site to make the molecule more polarised (or electrically charged) and thus more able to be excreted by the body.
Phase I by definition creates a potentially toxic reactive oxygen intermediate with free radical activity, for which endogenous antioxidant protection is required. This phase is launched primarily by the cytochrome P450 enzyme family whose role is, like the antibody system, to recognise and initiate the biotransformation of a wide diversity of substrates. Excessive phase I activity can generate more dangerous toxic complications. This might happen, for example, in fasting or other sudden loss of adipose tissue where fat-soluble metabolites are stored. The same challenge may occur if there is a reduction in antioxidant capacity, such as in heavy smoking, exposure to industrial pollutants (which can both induce excessive phase I activity as well) or chronically deficient diets. Its role in oestrogen metabolism is a particularly good example of the liver’s significance in health and disease. Unlike other hormones, oestrogen is eliminated by a full phase I and II mechanism, suggesting that the body evolved to see it as a threat (oestrogen is one of the most powerful of the body’s major hormones). The rate of phase I detoxification (through cytochromes P450: CYP1A1, CYP1B1, and CYP3A4; see also Chapter 5) is determined by many factors, including individual ‘mini-mutations’ of P450s known as single nucleotide polymorphisms. As phase I metabolites can have anti-oestrogenic or DNA-damaging effects, then the impact of these variations on the incidence of breast uterine and prostate cancer is known to be substantial.2 The cancer-protective role of cruciferous vegetables (through glucosinolates) is likely to include a positive effect on these detoxification processes (see also Chapter 2).3
Phase II reactions should cut in to neutralise the transformed substrates quickly, to reduce toxic burdens and relieve antioxidant defences. They involve conjugation with glucuronic acid, sulphates, glutathione, glycine or other amino acids, acetylation or methylation. These protective processes are known to be augmented by a diet high in flavonoids, such as fruit and vegetables,4,5 particularly cruciferous vegetables,6 onions, red grapes and soy.7 The onset of diseases, notably cancer, has been often linked to an imbalance between phase I and phase II and the protective benefits of fruit and vegetables against cancer have been put down to this effect. Fundamentally, it is now understood that many of the phytochemicals in fruits and vegetables activate the Nrf2/ARE pathway and thereby induce the production of phase II and antioxidant enzymes. (ARE stands for ‘antioxidant response element’.)
Liver remedies may help reduce toxic burdens by shifting the phase I/II balance towards the latter, stimulating cleansing bile flow (see previous section) or by directly protecting the liver tissue itself from oxidant attack. Excessive or phase I or cytochrome P450 activity may lead to mutation, cancer or tissue necrosis, particularly of the liver with its particularly high content of cytochrome P450.8
Consequently, the liver is an extremely active organ in the metabolism of ingested materials and it is known to be particularly vulnerable to the effects of agents that are not readily metabolised or excreted. These are known to include many modern drugs, agrochemicals and other modern environmental pollutants. Although severe hepatic damage is not common, constant exposure to new industrial agents, particularly if at relatively high levels and when combined with other well-known burdens on the liver, alcohol and high fat diets, can lead to a modern syndrome of functional liver distress.
Because of the central role of the liver in immunological function and detoxification, there may also be a significant population of patients, perhaps presenting with other conditions (notably skin and bowel diseases, chronic allergic states, autoimmune disease and other chronic inflammatory disorders), in whom subtle liver damage or dysfunction may already have occurred and can be contributing further to the wider pathology. The increased incidence of viral hepatitis is both the major cause of long-term hepatic disorders and a likely added complication of some of the above adverse changes. The mechanisms of chronic liver damage may include immunological disturbances such as chronic active hepatitis and primary biliary cirrhosis, or may be more subtle with perhaps minor alterations in liver function tests or merely subjective intolerance of fats and alcohol (see Poor liver function below).
As hepatic damage usually starts with classic pathological processes of fatty infiltration, in theory a good liver remedy should have appropriate antioxidant properties. As these need to be focused in liver tissue, they should also be readily absorbed, require little extra liver metabolism and preferably should be retained in the enterohepatic circulation. The best such herbs will contain phytochemicals that activate the Nrf2/ARE pathway, such as curcumin.
Many plants contain constituents that have been experimentally shown to have beneficial effects on liver cells, at least under laboratory conditions. Early research investigated the hepatoprotective effects of agents in models of liver toxicity (for example, carbon tetrachloride (CCl4), D-galactosamine, paracetamol and Amanita mushrooms) and it was such work that identified remedies such as Silybum marianum (St Mary’s thistle). This and more recent evidence more widely points to benefits on hepatic function of a diet rich in fruit and vegetables (as noted above). The case for traditional herbal practice in its particular focus on the liver as a centre-piece of therapeutics is also supported.
Particularly likely to feature in evidence are flavonoids and related polyphenols. For example, the ubiquitous flavonoid quercetin is one of a number of antioxidants that have been shown to reduce the high level of chromosomal aberrations found in cases of viral hepatitis associated with environmental pollution.9 Oral administration of the flavonoids daidzin, daidzein and puerarin, from the Chinese herb Pueraria lobata, was effective in lowering blood alcohol levels and shortened sleep time induced by alcohol ingestion.10,11
Glycyrrhizin, a major component of licorice, has been used intravenously for the treatment of chronic hepatitis B in Japan and improves liver function with occasional complete recovery from hepatitis.12 In vitro studies suggest a hitherto unique mechanism involving intracellular transport across Golgi membranes.13 In addition, weak binding activity to steroid receptors has been demonstrated.14 Derivatives of glycyrrhizinic acid promote a decrease in the rate of lipid peroxide oxygenation and in the inhibition of liver enzyme activity (ALT, AST) and also increase choleresis.15 There is evidence of a choleretic action,16 also involving glycyrrhizin. (See also the licorice monograph.)
Piperine, an active alkaloidal constituent of the extract obtained from Piper longum and P. nigrum, was evaluated for its antihepatotoxic potential against CCl4 and other agents, and was found to have an appreciable benefit, but at levels lower than for silymarin.17
The water extract from the root of Salvia miltiorrhiza showed a protective effect on cultured rat hepatocytes against CCl4-induced necrosis. Lithospermate B, a tetramer of caffeic acid, was isolated and found to be an active constituent. Lithospermate B was also found to have a potent hepatoprotective activity, not only in vitro but also for in vivo experimental liver injuries induced by CCl4 or D-galactosamine-lipopolysaccharide.18 Various other metabolites of caffeic acid formed in bap are likely to account for many beneficial effects in that organ; these metabolites include cyclolignan derivatives as oxidation products, ferulic and isoferulic acid as methylation products and aesculetin as a cyclisation product.19
The Phyllanthus genus has been known to contain a number of biologically active constituents such as an angiotensin-converting enzyme inhibitor and HIV reverse transcriptase inhibitors.20Phyllanthus amarus has shown antihepatitis B virus activity.21,22P. niruri from Malaysia contains various constituents with demonstrated effects aginst hepatitis B.23P. ussuriensis, a Korean native species, has been used to treat several infectious diseases, including hepatitis in folk medicine, and antihepatotoxic effects have been confirmed in vitro.24 It inhibited hepatitis B virus polymerase activity, decreased episomal hepatitis B virus DNA content and suppressed virus release into culture medium. A number of mechanisms have been elucidated.25
Silybum marianum (St Mary’s or milk thistle) seed, widely used in Europe for its hepatic reputation, contains the flavonol lignan complex silymarin with antifibrotic activity in diseased liver,26 and there are other effects observed in diseased liver,27 including reducing serum transaminases in patients with chronic viral hepatitis.28 The traditional liver remedy Schisandra chinensis contains constituents with antihepatitis activity,29 and has been shown to reduce circulating monocyte counts (although with no effects on other white blood cell counts) after 2 weeks’ treatment of human subjects infected with hepatitis B.30 (See more in the St Mary’s thistle monograph.)
The Chinese remedy Sophora flavescens has also been studies experimentally and has shown promising if not conclusive evidence of effectiveness in hepatitis B.31,32
Evidence of hepatoprotective effects also arises from studies on other traditional remedies: Artemisia spp.,33,34Azadirachta indica leaf extract,35Picrorrhiza kurroa,36Osbeckia octandra,37Gynostemma pentaphyllum,38Inula britannica L. subsp. japonica,39Salvia plebeia.40 Other plant hepatoprotectors have been reviewed.41–43
Schisandra has emerged as a key herb for boosting phase I and especially phase II detoxification processes by the liver. As stated previously, phase I and II reactions are involved in the metabolism of mainly xenobiotic substances by the liver. Phase I reactions (which involve cytochrome P450) can result in the production of a more toxic compound (a process called bioactivation). Despite being a postulated powerful inducer of phase I enzymes, Schisandra does not cause harmful bioactivation in vivo (e.g. after co-administration of paracetamol) and in vitro studies have indicated that constituents of Schisandra decrease the mutagenicity of benzo(alpha)pyrene by influencing hepatic metabolism.44,45 The probable reason is that Schisandra powerfully induces phase II enzymes as well, which results in the rapid clearance of the potentially toxic metabolite.46 Other keys herbs in this regard include turmeric (see the turmeric monograph), garlic, green tea, rosemary and broccoli sprouts (all influencing the Nrf2/ARE pathway). St Mary’s thistle is unlikely to exert clinically relevant effects on hepatic phase I/II detoxification processes, with its value being more linked to hepatoprotective activity (see below).
Another hitherto little explored hepatic activity may also be a feature of some herbal preparations: extracts of Thuja occidentalis and Echinacea purpurea have stimulated Kupffer cell phagocytosis in vitro.47
The liver has a central role in a wider range of defence mechanisms. For example, the Kupffer cells are a final screen between enteric antigens and the body cavities, various phase II liver enzymes such as glutathione-S-transferase (GST) are important detoxifiers of carcinogens, and the transport of IgA and IgA immune complexes by the liver from serum to bile may provide a host defence against enteric pathogens.48 These key functions raise the possibility that hepatic remedies might have wider applications.
For example, out of various spices and leafy vegetables screened for their influence on GST in Swiss mice, cumin seeds, poppy seeds, asafoetida, turmeric, neem flowers and basil leaves among other spices increased GST activity high enough in the stomach, liver and oesophagus to be considered as significantly contributing to protection against carcinogenesis.49 Several naturally occurring flavonoids and other polyphenols, prominently tannic acid, were also shown to exert varying degrees of concentration-dependent inhibition on uncharacterised rat liver GST.50 Sesquiterpenes such as beta-caryophyllene, beta-caryophyllene oxide, alpha-humulene, alpha-humulene epoxide I and eugenol, all found in cloves, have demonstrated significant activity as inducers of GST in the mouse liver and small intestine,51 as has myristicin, a major aromatic constituent of parsley.52 Of course, the most active liver phase II enzyme inducers are the brassica glucosinolates, and their protective properties against experimental carcinogenesis is well described (see Chapter 2). Another key example is turmeric (see the turmeric monograph).
Capsaicin inhibited the formation by liver microsomal fractions of all metabolites of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a potent carcinogen in tobacco and tobacco smoke. With other similar results, such findings suggest that it possesses antimutagenic and anticarcinogenic properties through the inhibition of xenobiotic metabolising enzymes.53
There are few guides to the use of hepatoprotectives (or hepatics) in conventional Western medicine. This is a concept more familiar in mainland Europe and further East, but has become a more popular concept with the marketing of St Mary’s thistle (milk thistle) seed.
Hepatoprotectives are remedies claimed to help reduce damage caused to the liver from hepatic stressors and disease. Given what has been said earlier, good hepatoprotectives are most likely to deliver antioxidant activity into liver hepatocytes and induce cellular protective mechanisms.
The most widely used hepatoprotectives in European phytomedicine appear to have few contraindications. They are generally mild treatments and, as antioxidants, may have wider benefits. To some extent they may merely extend the known beneficial effect on the liver of plant foods. Nevertheless, some of the remedies mentioned above may have individual characteristics that need consideration, especially in liver carcinoma.
Diseases with early morning symptoms may be associated with liver disturbances on the assumption that the liver has been particularly active overnight during sleep. The occurrence of such signs might in itself be an indication for the use of hepatoprotectives remedies.
Traditional enquiry might also elicit how well a patient handled alcohol and dietary fats. Although difficulties here might rather suggest the use of cholagogues and choleretics, hepatoprotectives would be particularly indicated if symptoms like headaches were associated with such consumption.
Hepatoprotectives are very often employed in the treatment of migraine, often seen traditionally as a liver condition.
Western practitioners are reminded of the ancient Greek humoral concept of ‘black bile’ (lit: melancholia) and the early view of clinical depression as a somatic rather than a psychological phenomenon. Using hepatoprotectives and other treatments for liver and digestive function as part of a fresh approach to the management of depression can be surprisingly productive in some cases.
Hepatoprotectives are best applied before breakfast in the morning and before the last meal of the day, to take account of the extra liver activity during the night. However, they may also be taken before meals throughout the day.
Long-term therapy is quite appropriate for those such as St Mary’s thistle and artichoke, which have established their safety from extensive human use.
Hepatoprotectives may also be usefully applied in some cases (depending on other factors) of:
There is also a strong case for their use in anticipation of, or association with, the prescription of powerful chemotherapeutic agents in, for example, the treatment of tuberculosis, cancer, psychoses and frequent recourse to general anaesthetics (see also Chapter 8).
For evidence of adverse effects of herbal products on the liver, see Chapter 5.
The most important causes of acute hepatitis are the hepatitis A, B and C viruses. Other viruses, including Epstein-Barr virus and cytomegalovirus, may also cause acute hepatitis. All these viruses (except A) have a viral envelope and hence may have some susceptibility to Hypericum perforatum (St John’s wort), which is active against enveloped viruses.
Acute hepatitis can be treated using herbal medicine. In the case of hepatitis A, treatment can lead to rapid recovery and protection against post-hepatitis syndrome. For hepatitis B and C, herbal treatment will mainly help to prevent the disease becoming chronic. The small amounts of alcohol involved from using extracts and tinctures will not be a problem to most patients. However, if there is a difficulty with alcohol then tablets, capsules, glycerol extracts and/or infusions or decoctions can be prescribed. As with other acute infections, oral herbal treatment should not be administered while there is vomiting.
It is hoped that individual strategies will be devised for each patient, taking some of the insights in this and other chapters into account, but essential aspects of treatment are as follows:
• Diaphoretics are indicated in all acute infections accompanied by fever. These include Tilia (lime flowers), Sambucus (elder) and Achillea (yarrow) and are best taken as an infusion. Diaphoretics are assisted by combination with a diffuse stimulant such as Zingiber (ginger)
• Antiviral agents, which for hepatitis include Hypericum, Phyllanthus and Thuja
• Immune-enhancing herbs, especially Echinacea and Andrographis
• Hepatoprotective agents to minimise liver damage such as Silybum, Bupleurum, Taraxacum radix (dandelion root), Cynara (globe artichoke) and Andrographis
• Post-hepatitis syndrome should mainly be treated with the hepatoprotectives listed above.
It should be noted that higher doses need to be employed for acute cases (compared to treating chronic conditions).
Chronic viral hepatitis or chronic persistent hepatitis usually results from infection with hepatitis B or C. Especially with hepatitis C, some features of the disease may resemble autoimmune hepatitis. (Frank autoimmune liver disease is known as chronic active hepatitis. A general treatment approach for autoimmune disease is outlined in Chapter 8.)
Treatment of chronic viral hepatitis shares many similarities with the treatment of acute viral hepatitis. Essential features of treatment are:
• immune-enhancing agents such as Echinacea, Andrographis and Astragalus for any chronic infection
• hepatoprotective agents as described previously and particularly Schisandra and Silybum in a more concentrated form such as tablets or capsules containing the silymarin extract
• antiviral agents (see above). The use of Phyllanthus may be suitable for chronic viral hepatitis (see previous) as is St John’s wort
• herbal antioxidants/cytoprotectives to provide further protection for hepatocytes, such as turmeric and green tea.
Case history
A female patient aged 56 years had been diagnosed with chronic hepatitis C 2 years prior. Her genotype (genotype I) was less responsive to interferon therapy, although it had been trialled for 1 year. ALT and AST were moderately elevated at the time of her first consultation (130 and 95 U/L, respectively), although they were severely elevated at the time of diagnosis. Over the 8 years of treatment with phytotherapy her condition has progressively improved. There have been no acute episodes and her ALT and AST readings are nearly normal at 47 and 53 U/L, respectively. This result was achieved with no other additional therapy and the patient remains otherwise well.
Treatment consisted of tablet products delivering the following daily doses:
Astragalus | 3.4 g |
Echinacea purpurea root | 2.6 g |
Grape seed extract | 100 mg |
Turmeric | 4 g |
Green tea extract | 333 mg |
Rosemary | 2 g |
Siberian ginseng | 3 g |
St John’s wort (from standardised extract) | 5.4 g |
Schisandra | 2 g |
Silymarin extract (from St Mary’s thistle) | 630 mg |
In cirrhosis, widespread death of liver cells, which can result from many causes but is most commonly due to alcohol abuse, is accompanied and followed by progressive fibrosis and distortion of liver architecture. Since alcohol is forbidden, herbal treatment should be in the form of tablets, capsules, glycerol extracts, infusions and/or decoctions. The main herbal treatment is based around hepatic trophorestoratives, especially concentrated tablets of Silybum marianum. Other important herbs in this category include Schisandra, Taraxacum radix (dandelion root), Cynara (globe artichoke), Bupleurum and Andrographis. Berberine-containing herbs can also be indicated (see the Berberis monograph). Although cirrhosis is a progressive disease, the rate of progression varies and the outlook is related to many factors. In this context, herbal treatment can make a significant positive contribution.
Antifibrotic activity is another area of herbal activity relevant to cirrhosis that is attracting research interest. The laying down of excessive fibrotic tissue in response to repeated liver damage is the main disruptive pathological change in cirrhosis. Centella asiatica (gotu kola) is a herb with potential to reduce an excessive fibrotic response (see gotu kola monograph). Salvia miltiorrhiza (dan shen) is also showing promising activity in this regard.54,55
The liver plays a vital role in detoxification and many other metabolic processes in the body. Phytotherapists and naturopaths recognise a condition where liver function is below optimum, although no medically observable liver disease or liver damage may be present. Because of the importance of the liver, a poorly functioning liver can have a wide-ranging impact on health.
Symptoms that may be due to poor liver function include sluggish digestion, fat intolerance, nausea, chronic constipation and chemical, food or drug intolerances. A poorly functioning liver may also contribute to a number of disease states such as psoriasis, autoimmune disease, irritable bowel syndrome, allergies and cancer. Patients might reveal a history of past liver infection, infestation or damage, alcohol or drug abuse, or exposure to medical drugs or environmental pollutants such as pesticides and/or have abdominal obesity. Drug side effects are more likely to occur in patients with poor liver function.
Depending on the symptoms, treatment is based on the following:
• Hepatoprotective and hepatic trophorestorative herbs, especially if there is a history of liver damage or exposure to toxins. Principal herbs include Silybum, Cynara and Taraxacum. Schisandra is particularly useful since it also enhances the detoxifying capacity of the liver (see above). These herbs will assist in cases of nausea and intolerances from any cause
• Choleretic herbs to boost liver function are particularly indicated if digestive symptoms are predominant. They will also boost detoxification via bile and therefore can be valuable in conditions such as psoriasis and cancer. Most of the hepatoprotective herbs listed above have a gentle choleretic activity, but strongly choleretic herbs include Hydrastis (golden seal), Berberis vulgaris (barberry), Chelidonium and bitter herbs. These strongly choleretic herbs tend to cause nausea and irritability in a patient who has some history of liver damage. They should therefore be avoided at first in these circumstances and only introduced after prior treatment with the hepatic trophorestoratives noted above
• Depurative herbs are also indicated in cases where hepatic detoxification may be inadequate. Those which act principally via the liver and digestion include Arctium (burdock), Rumex crispus (yellow dock) and Fumaria (fumitory)
• Other herbs (in addition to Schisandra) that specifically boost phase II hepatic detoxification should also be considered, including broccoli sprouts, rosemary, turmeric, garlic and green tea.
Case history
A female patient aged 38 wished to take the contraceptive pill. She found that even a low-dose pill still caused symptoms of female hormone excess such as abdominal bloating, weight gain, nausea and depression. She had a past history of liver damage due to hydatid worm cysts during childhood.
Silybum marianum | 1:1 | 30 mL |
Taraxacum officinale radix | 1:2 | 35 mL |
Schisandra chinensis | 1:2 | 35 mL |
total | 100 mL |
Dose: 5 mL with water twice a day.
The patient found she could take the pill without any adverse effects as long as she also took the herbal treatment.
References
1. Dunning AM. Polymorphisms associated with circulating sex hormone levels in postmenopausal women. J Natl Cancer Inst. 2004;96:936–945.
2. Yager JD. Endogenous estrogens as carcinogens through metabolic activation. J Natl Cancer Inst Monogr. 2000;27:67–73.
3. Bonnesen C, Stephensen PU, Andersen O, et al. Modulation of cytochrome P-450 and glutathione S-transferase isoform expression in vivo by intact and degraded indolyl glucosinolates. Nutr Cancer. 1999;33(2):178–187.
4. Moon YJ, Wang X, Morris ME. Dietary flavonoids: effects on xenobiotic and carcinogen metabolism. Toxicol In Vitro. 2006;20(2):187–210.
5. van Breda SG, van Agen E, Engels LG, et al. Altered vegetable intake affects pivotal carcinogenesis pathways in colon mucosa from adenoma patients and controls. Carcinogenesis. 2004;25(11):2207–2210.
6. Riedl MA, Saxon A, Diaz-Sanchez D. Oral sulforaphane increases phase II antioxidant enzymes in the human upper airway. Clin Immunol. 2009;130(3):244–251.
7. Appelt LC, Reicks MM. Soy feeding induces phase II enzymes in rat tissues. Nutr Cancer. 1997;28(3):270–275.
8. Pessayre D. Cytochromes P450 and formation of reactive metabolites. Role in drug induced hepatotoxicity. Therapie (Paris). 1993;48(6):537–548.
9. Peresadin NA, Frolov VM, Pinskii LL. Correction with antioxidants of cytogenetic disturbances in viral hepatitis. Lik Sprava. 1995;1–2:76–79.
10. Lin RC, Guthrie S, Xie CY, et al. Isoflavonoid compounds extracted from Pueraria lobata suppress alcohol preference in a pharmacogenetic rat model of alcoholism. Alcohol Clin Exp Res. 1996;20(4):659–663.
11. Xie CI, Lin RC, Antony V, et al. Daidzin, an antioxidant isoflavonoid, decreases blood alcohol levels and shortens sleep time induced by ethanol intoxication. Alcohol Clin Exp Res. 1994;18(6):1443–1447.
12. Sato H, Goto W, Yamamura J, et al. Therapeutic basis of glycyrrhizin on chronic hepatitis B. Antiviral Res. 1996;30(2–3):171–177.
13. Takahara T, Watanabe A, Shiraki K. Effects of glycyrrhizin on hepatitis B surface antigen: a biochemical and morphological study. J Hepatol. 1994;21(4):601–609.
14. Tamaya T, Sato S, Okada H. Inhibition by plant herb extracts of steroid bindings in uterus, liver and serum of the rabbit. Acta Obstet Gynecol Scand. 1986;65(8):839–842.
15. Nasyrov KhM, Chepurina LS, Kireeva RM. Study of hepatoprotective and choleretic activity of glycyrrhizinic acid derivatives. Eksp Klin Farmakol. 1995;58(6):60–63.
16. Raggi MA, Bugamelli F, Nobile L, et al. The choleretic effects of licorice: identification and determination of the pharmacologically active components of Glycyrrhiza glabra. Boll Chim Farm. 1995;134(11):634–638.
17. Koul IB, Kapil A. Evaluation of the liver protective potential of piperine, an active principle of black and long peppers. Planta Med. 1993;59(5):413–417.
18. Hase K, Kasimu R, Basnet P, et al. Preventive effect of lithospermate B from Salvia miltiorhiza on experimental hepatitis induced by carbon tetrachloride or D-galactosamine-lipopolysaccharide. Planta Med. 1997;63(1):22–26.
19. Gumbinger HG, Vahlensieck U, Winterhoff H. Metabolism of caffeic acid in the isolated perfused rat liver. Planta Med. 1993;59(6):491–493.
20. Munshi A, Mehrotra R, Ramesh R, Panda SK. Evaluation of anti-hepadnavirus activity of Phyllanthus amarus and Phyllanthus maderaspatensis in duck hepatitis B virus carrier Pekin ducks. J Med Virol. 1993;41(4):275–281.
21. Liu J, Lin H, McIntosh H. Genus Phyllanthus for chronic hepatitis B virus infection: a systematic review. J Viral Hepat. 2001;8(5):358–366.
22. Martin KW, Ernst E. Antiviral agents from plants and herbs: a systematic review. Antivir Ther. 2003;8(2):77–90.
23. Bagalkotkar G, Sagineedu SR, Saad MS, Stanslas J. Phytochemicals from Phyllanthus niruri Linn. and their pharmacological properties: a review. J Pharm Pharmacol. 2006;58(12):1559–1570.
24. Moon Y-S, Lim WS, Lee MK, et al. Antihepatotoxic effect of callus cultures of the P. ussuriensis. Seoul Univ J Pharm Sci. 1995;20:21–31.
25. Lee C-D, Ott M, Thyagarajan SP, et al. Phyllanthus amarus down-regulates hepatitis B virus mRNA transcription and replication. Eur J Clin Invest. 1996;26(12):1069–1076.
26. Boigk G, Stroeter L, Waldschmidt J, et al. Silymarin retards collagen accumulation in rat secondary biliary fibrosis. J Hepatol. 1995;23(suppl 1):142.
27. Tamayo C, Diamond S. Review of clinical trials evaluating safety and efficacy of milk thistle (Silybum marianum [L.] Gaertn.). Integr Cancer Ther. 2007;6(2):146–157.
28. Mayer KE, Myers RP, Lee SS. Silymarin treatment of viral hepatitis: a systematic review. J Viral Hepat. 2005;12(6):559–567.
29. Liu GT. Pharmacological actions and clinical use of fructus schizandrae. Chin Med J (Engl). 1989;102(10):740–749.
30. Yip AY, Loo WT, Chow LW. Fructus Schisandrae (Wuweizi) containing compound in modulating human lymphatic system – a phase I minimization clinical trial. Biomed Pharmacother. 2007;61(9):588–590.
31. Liu J, Zhu M, Shi R, Yang M. Radix Sophorae flavescentis for chronic hepatitis B: a systematic review of randomized trials. Am J Chin Med. 2003;31(3):337–354.
32. Long Y, Lin XT, Zeng KL, Zhang L. Efficacy of intramuscular matrine in the treatment of chronic hepatitis B. Hepatobiliary Pancreat Dis Int. 2004;3(1):69–72.
33. Janbaz KH, Gilani AH. Evaluation of the protective potential of Artemisia maritima extract on acetaminophen- and CC14-induced liver damage. J Ethnopharmacol. 1995;47(1):43–47.
34. Yin J, Wennberg RP, Miller M. Induction of hepatic bilirubin and drug metabolizing enzymes by individual herbs present in the traditional Chinese medicine, yin zhi huang. Dev Pharmacol Ther. 1993;20(3–4):186–194.
35. Chattopadhyay RR, Sarkar SK, Ganguly S, et al. Hepatoprotective activity of Azadirachta indica leaves on paracetamol induced hepatic damage in rats. Indian J Exp Biol. 1992;8:738–740.
36. Dhawan BN. Picroliv – a new hepatoprotective agent from an Indian medicinal plant, Picrorrhiza kurroa. Med Chem Res. 1995;5(8):595–605.
37. Thabrew MI, Hughes RD, Gove CD, et al. Protective effects of Osbeckia octandra against paracetamol-induced liver injury. Xenobiotica. 1995;9:1009–1017.
38. Li L, Jiao L, Lau BH. Protective effect of gypenosides against oxidative stress in phagocytes, vascular endothelial cells and liver microsomes. Cancer Biother. 1993;8(3):263–272.
39. Iijima K, Kiyohara H, Tanaka M, et al. Preventive effect of taraxasteryl acetate from Inula britannica subsp. Japonica on experimental hepatitis in vivo. Planta Med. 1995;61(1):50–53.
40. Lin C-C, Lin J-K, Chang C-H. Evaluation of hepatoprotective effects of ‘Chhit-Chan-Than’ from Taiwan. Int J Pharmacog. 1995;33(2):139–143.
41. Rumyantseva ZhN, Gudivok YS. Search for hepatoprotectors among preparations of plant origin. Rastitel’nye Resursy. 1993;29(1):88–97.
42. Rakhmanin YA, Kushnerova NF, Gordeichuk TN, et al. Metabolic responses of the liver exposed to carbon tetrachloride and their correction with plant antioxidants. Gig Sanit. 1997;1:30–32.
43. Negi AS, Kumar JK, Luqman S, et al. Recent advances in plant hepatoprotectives: a chemical and biological profile of some important leads. Med Res Rev. 2008;28(5):746–772.
44. Liu KT, Cresteil T, Columelli S, et al. Pharmacological properties of dibenzo[a,c]cyclooctene derivatives isolated from Fructus schizandrae chinensis. II. Induction of phenobarbital-like hepatic monooxygenases. Chem Biol Interact. 1982;39(3):315–330.
45. Liu KT, Lesca P. Pharmacological properties of dibenzo[a,c]cyclooctene derivatives isolated from Fructus schizandrae chinensis. I. Interaction with rat liver cytochrome P-450 and inhibition of xenobiotic metabolism and mutagenicity. Chem Biol Interact. 1982;39(3):301–314.
46. Bone K. Schisandra – the complete liver herb. Townsend Lett Doctors Patients. 2003;245:108–112.
47. Vömel T. Effect of a plant immunostimulant on phagocytosis of erythrocytes by the reticulohistiocytary system of isolated perfused rat liver. Arzneimittel-Forschung. 1985;35(9):1437–1439.
48. Kleinman RE, Harmatz PR, Walker WA. The liver: an integral part of the enteric mucosal immune system. Hepatology. 1982;2(3):379–384.
49. Aruna K, Sivaramakrishnan VM. Plant products as protective agents against cancer. Indian J Exp Biol. 1990;11:1008–1011.
50. Zhang K, Das NP. Inhibitory effects of plant polyphenols on rat liver glutathione S-transferases. Biochem Pharmacol. 1994;47(11):2063–2068.
51. Zheng GQ, Kenney PM, Lam LK. Sesquiterpenes from clove (Eugenia caryophyllata) as potential anticarcinogenic agents. J Nat Prod. 1992;55(7):999–1003.
52. Zheng GQ, Kenney PM, Zhang J, Lam LK. Inhibition of benzo[a]pyrene-induced tumorigenesis by myristicin, a volatile aroma constituent of parsley leaf oil. Carcinogenesis. 1992;13(10):1921–1923.
53. Miller CH, Zhang Z, Hamilton SM, Teel RW. Effects of capsaicin on liver microsomal metabolism of the tobacco-specific nitrosamine NNK. Cancer Lett. 1993;75(1):45–52.
54. Tao YY, Liu CH. Progress of research on mechanism of Salvia miltiorrhiza and its chemical ingredients against liver fibrosis. Zhong Xi Yi Jie He Xue Bao. 2004;2(2):145–148.
55. Stickel F, Brinkhaus B, Krähmer N, et al. Antifibrotic properties of botanicals in chronic liver disease. Hepatogastroenterology. 2002;49(46):1102–1108.
Cardiovascular system
Apart from their use to provide non-specific support for recuperation and repair, specific phytotherapeutic strategies include the following.
• chronic, non-severe hypertension
• atheromatous cardiovascular conditions
Because of its use of secondary plant products, particular caution is necessary in applying phytotherapy in cases of:
A phytotherapeutic perspective on the circulatory system has to take two parts. Most of what modern medicine understands of the system arises from preoccupation with disease states such as hypertension, hypercholesterolaemia, atherosclerosis, clotting disturbances and thrombosis, and coronary diseases that were barely understood in an earlier era and for which traditional physicians cannot have developed therapeutic strategies. There can be no basis therefore for directly applying old treatments to the new conditions. Nevertheless, as this chapter will show, herbal remedies show considerable promise across many of these modern conditions and their use for this purpose, at least in Europe, has been considerably modified compared with their traditional indications. Three of the best-selling herbal products in Europe, Ginkgo, garlic and Crataegus (hawthorn), are traditional remedies highly adapted to new and productive ends.
Nevertheless, to understand more effectively the potential of medicinal plants in affecting circulatory functions, an appreciation of the earlier traditional perspective will be helpful. It is immediately obvious that before modern instruments, human experience of the circulatory system and the effects of treatments upon it were very different. As shall be seen, the insights developed in these early times usefully inform modern prospects.
The circulatory apparatus of William Harvey provided a mechanistic framework for modern advances that was, however, of little application to everyday practice in his time. The common experience was that there was vital movement within the body, as measured by pulse, heartbeat and breathing, and that there was a red fluid whose presence was clearly essential. Even now microscopic film of tissue circulation resonates uncannily with the beats of tribal music! It was relatively easy to link this with the main manifestation of moving blood: the heat of the living body and the variations in that heat in health and disease. In short, blood pulsated and warmed and was generally linked with the common speculation that there must be circulation of energies, fluids and nutrients around the body. Circulation was marked by:
The heart was obviously associated with all this, but as much as a resonator with the vital pulse as its director. It was the wider pulse itself (reflected in the driving rhythms of early tribal music) that was important in early experience of the circulatory system; it was clearly linked to wider vital events: activity, excitement, emotional stimulation and, in medicine, notably the fever.
Therapeutics in fever focused on dispersing agents to distribute excessive heat and circulation and (as ‘diaphoretics’) to diffuse the poisons clearly involved through the sweat glands. The detoxifying theme recurred in many traditional concepts of ‘blood poisons’ as a cause of inflammatory diseases, and the use of ‘blood cleansers’ or ‘blood purifiers’, often very vigorously, to treat them.
Also requiring eliminatives (mainly diuretics and laxatives) was oedema, one of the most common indications of poor circulation in the past. As briefly elaborated in Chapter 1, the traditional perspective closely linked circulatory function with elimination.
Traditional views also linked circulation with the assimilation and processing of nutrients. The vital pulse and heat were weakened in debility and exhaustion, conditions associated with coldness and pallor. The main treatments were in effect ‘blood tonics’, warming nutrients (often since found to be rich in mineral nutrients).
There will be profit in revisiting these perspectives in developing modern strategies for the treatment of circulatory problems using herbal remedies. This is even more justified when the phenomenon of circulation itself is reviewed.
Where there has been speculation about the nature of the circulatory system in traditional medicine, it has tended to infer broad currents rather than Harvey’s route map. The closest to the latter were the meridians of Chinese medicine, although even these were speculative phenomena not associated with anatomical conduits.1 The phenomenological perspective of tradition turns out, however, to be closer to the reality than the conventional understanding of arteries, veins and capillaries might suggest.
As far as most tissue cells are concerned, blood flow is not through vessels at all. When plasma filters through the capillary walls to bathe the tissues, it does not diffuse freely. In most tissues, cells are embedded in a gelatinous matrix, formed of complexes of hyaluronic acid which is largely impermeable to aqueous fluids. Movement of plasma is thus confined through clefts and cleavages in the matrix. The interstitial matrix thus both maintains tissue integrity and restricts the free flow of the circulation; oedema is the main symptom of breakdown in this important construct.
The effect of the interstitial matrix on circulatory dynamics is profound. As far as tissues are concerned, circulation is not a Harveyian affair at all, it is more a diffusive process marked by local and wider ‘oceanic’ currents. Factors that affect tissue circulation are thus different from those that preoccupy modern cardiovascular medicine. Atherosclerosis and thrombosis cause serious local circulatory harm, of course, but they impact on general circulation only when very severe. More important for circulatory health in the tissues are such factors as capillary wall integrity, the local responses to local environmental changes of powerful vasoactive agents such as the kinins and histamine, venous or lymphatic stasis or congestion with subsequent oedema and toxicity.
A glance at any pathology text will confirm that the cellular processes of disease are remarkably consistent. Pathological deterioration starts with biochemical lesions, then a variety of stages supervene including intracellular lesions, cell hypertrophy and a range of possible degenerative changes, cellular swelling due to water influx into the cell, fatty change or accumulation, atrophy, necrosis, possibly leading to inflammation or calcification. Most detectable disease states in the body are classified by one or more of these processes. Atherosclerosis, for example, involves fatty infiltration and then calcification of the tissues in the arterial walls.
Moreover, the very first initiating trauma is even more consistent. The most likely first step in tissue damage is a relative deficiency of oxygenated blood and fluids. Physical injury is the most likely initiating trauma followed by an accumulation of external or endogenous toxic substances. In both the first and last cases poor tissue perfusion is critical. There are a number of ways in which tissue circulation can be interrupted but there is a clear prima facie case for maintaining tissue perfusion as a core disease-preventing strategy.
One of the most fascinating prospects for the revival of traditional herbal and dietary approaches is in the number of ways in which plant constituents, such as flavonoids, anthocyanins, sesquiterpenes and pungent principles, appear to act beneficially on local circulatory processes.
It is a consistent theme throughout history that the ‘heating’ remedies were literally life enhancing (see p. 4 and p. 9). The pungent remedies such as cayenne, ginger and raw garlic had reputations that transcended the merely mundane. It is known that they do increase tissue perfusion and blood flow. Everyday subjective experiences of increased body heat after eating spicy food can be confirmed with thermometers. Reference to the ginger monograph reveals a number of studies demonstrating a thermogenic effect, involving such mechanisms as increased catecholamine production and cytokine activity. Supplementing rats’ diet with garlic powder increased rectal temperatures, blood noradrenaline levels and mitochondrial activity in brown adipose tissues, an activity that was inhibited by beta-adrenergic blockers.2
The prospects for closer investigation are intriguing. It is most probable that the traditional remedies most often used for their tissue warming benefits will show really exciting properties in the treatment of, or prophylaxis against, a range of degenerative diseases that may include atherosclerosis and other cardiovascular diseases. The fact that most are common ingredients of the diet adds even more to this project.
A promise of what may be in store for cayenne, ginger, cinnamon, turmeric and the like is the remarkable story of garlic, now possibly the most intensively studied of all medicinal plants and foods.
The chemistry of Allium sativum is complex and the multitude of garlic products available in the marketplace reflects this complexity.3 These types of preparations can be divided into three main groups, to which is added fresh garlic:
1. Carefully dried garlic powder that preserves the compound alliin (S-allylcysteine sulphoxide) and the enzyme alliinase. On disintegration of tablets or capsules containing this powder in the digestive tract, alliin comes into contact with alliinase and is converted to allicin. This must take place outside the stomach, as gastric acid inhibits alliinase. Enteric-coating of the tablets or capsules is therefore necessary. (This process mimics the chemical reaction that occurs when a fresh clove of garlic is crushed.) Allicin is unstable and breaks down further into compounds such as diallyl sulphides, ajoene and the vinyl dithiins (the metabolic pathways for allicin in the human body are not fully understood).
2. Aged garlic extracts or ‘odourless’ garlic products that are produced by a fermentation process. These preparations contain modified sulphur compounds such as S-allylcysteine.
3. Steam-distilled preparations of garlic (garlic oil) rich in diallyl sulphides.
Most of the published clinical studies on garlic have used ‘garlic powder’ preparations, although trials on aged garlic extracts, fresh garlic and garlic oil are also in the literature.
Many studies have demonstrated the lipid-lowering effects of garlic and the results of meta-analyses have supported the premise that garlic acts as a lipid-lowering agent. One examined five selected clinical trials on various garlic preparations with a total of 410 patients.4 The authors concluded that the best available evidence suggests that garlic, in an amount approximating one half to one clove per day, decreased total serum cholesterol levels by about 9%. About a year later a second meta-analysis was published by Silagy and Neil.5 These scientists included 16 clinical trials with a total of 952 patients. Again, a variety of garlic preparations were included in the meta-analysis. They found that garlic lowered cholesterol levels by 12% and that dried garlic powder preparations also lowered serum levels of triglycerides. In spite of some published negative trials (for example, on garlic6,7 and on garlic oil8), the latest meta-analysis taking 29 clinical trials up to 2007 found garlic significantly reduced total cholesterol and triglycerides, though exhibited no significant effect on LDL or HDL.9 The results of negative trials are confounded by the observation of Lawson and team that products used in clinical trials where garlic did not lower cholesterol often did not effectively release allicin.10
Perhaps the real value of garlic in the prevention and treatment of cardiovascular disease lies elsewhere. For example, a double blind, placebo-controlled study on 23 patients found that garlic powder tablets reduced the atherogenicity of low-density lipoprotein.11 In a controlled retrospective study on 202 healthy adults, divided equally between those taking garlic powder and controls, in which measures of the elastic properties of the aorta were used, garlic reduced age-related increases in aortic stiffness.12
An important trial looked at the effect of garlic powder intake over 4 years on arterial plaque. The trial was a randomised, double blind, placebo-controlled design involving 152 patients. Plaque volumes in both carotid and femoral arteries were measured by ultrasound. The increase in plaque volume over time was significantly reduced by garlic and in some cases there was a slight regression. The authors were accused of scientific fraud, but subsequently vindicated.13–15
Researchers from Germany report that, in test tubes, garlic prevents formation of ‘nanoplaques’ that can accumulate to cause arteriosclerosis. During a National Institutes of Health workshop on herbs and cardiovascular disease held in Bethesda, MD, in August 2002, Dr Günter Siegel from the Free University of Berlin, described his team’s research, which pinpoints exactly how garlic blunts plaque formation.16 In the presence of calcium, low-density lipoprotein (LDL)-cholesterol binds with molecules secreted from the inner lining of the arteries, forming tiny plaques that can accumulate and harden. HDL-cholesterol inhibits this process by absorbing excess plaque-forming molecules. Siegel’s team found that garlic extract works exactly the same way, but more potently. Garlic extract was two and a half times more effective in inhibiting plaque formation than was HDL-cholesterol.16 This has led to Siegel describing this form of garlic as phyto-HDL, that is a herb acting in the same beneficial way as HDL.
A meta-analysis of eight clinical trials (415 patients), all using the same garlic powder preparation, found that garlic caused a modest but significant reduction in both systolic and diastolic blood pressures.17 However, only three of the trials were specifically conducted in hypertensive patients and many had other methodological shortcomings. A review of aged garlic extract has assigned US National Health and Medical Research Council levels of evidence III-1 ratings on conclusions that 7.2 g has been associated with anti-clotting (in vivo studies), as well as modest reductions in blood pressure (an approximate 5.5% decrease in systolic blood pressure).18 In a recent meta-analysis involving ten trials, garlic reduced systolic blood pressure by 16.3 mmHg (95% CI 6.2 to 26.5) and diastolic pressure by 9.3 mmHg (95% CI 5.3 to 13.3) compared with placebo, although only in patients with elevated systolic blood pressure.19 Another contemporary meta-analysis of eleven studies showed a mean decrease of 4.6±2.8 mmHg for systolic blood pressure in the garlic groups compared to placebo (n=10; p=0.001), while the mean decrease in the hypertensive subgroup was 8.4±2.8 mmHg for systolic (n=4; p<0.001), and 7.3±1.5 mmHg for diastolic blood pressure (n=3; p<0.001).20
A platelet-inhibiting effect has been described for garlic. In a double blind, placebo-controlled study involving 60 volunteers with elevated cerebrovascular risk factors and increased spontaneous platelet aggregation, it was demonstrated that 800 mg/day of garlic powder over 4 weeks led to a significant reduction in platelet aggregation and circulating platelet aggregates.21 This inhibition of platelet aggregation by garlic powder was confirmed by another research group.22 However, the confounding issue of the various dosage forms of garlic was highlighted by a study of an oil extract of garlic, which found no significant effect on platelet aggregation.23 In contrast, consumption of a fresh clove of garlic daily for a period of 16 weeks reduced serum thromboxane by about 80%.24
One of the compounds responsible for the antiplatelet effect of garlic powder could be ajoene.25 This compound inhibits aggregation induced by all known platelet agonists in all species studied and prevents the amplification of platelet responses. Unlike aspirin, it acts by modifying the platelet membrane structure.
A review of published studies found that garlic consistently increased fibrinolytic activity after single or multiple doses. Garlic oil and garlic powder were both active, sometimes after only a single dose. The average increase in the reviewed studies was 58%.26 A 1991 controlled study using raw garlic demonstrated a significant increase in clotting time and fibrinolytic activity after 2 months in normal volunteers.27
In a randomised, placebo-controlled, double blind, crossover study in ten healthy volunteers, a single dose of 600 mg of garlic powder significantly reduced haematocrit (p<0.001), plasma viscosity (p<0.05) and plasma fibrinogen (p<0.05).28 Fibrinolytic activity was also significantly increased (p<0.01). A similar study design also found that a single 900 mg dose of garlic powder significantly increased capillary skin perfusion by 55% (p<0.01).29 Another study found that garlic powder (600 mg/day) administered for 7 days increased calf blood flow by approximately 15% (p=0.001).30
Two naturally produced gaseous signalling molecules play a key role in the regulation of cardiovascular physiology.31 These gaseous messengers, nitric oxide (NO) and carbon monoxide (CO), are synthesised by endogenous enzyme systems. Extensive research has shown that agents that improve their production protect the brain and heart against cardiovascular diseases, although this is sometimes controversial.31
In an intriguing scientific breakthrough, research on garlic has revealed it is involved in the generation of a significant third gaseous signalling molecule, namely hydrogen sulphide (H2S).32 H2S, also known as rotten egg gas, is toxic in high amounts. But it appears that the small quantities induced by the ingestion of garlic could play a role in its cardiovascular benefits, as well as the characteristic garlic breath. Scientists were able to demonstrate that garlic-derived organic polysulphides, such as diallyl disulphide and diallyl trisulphide, act as H2S donors.32 Human red blood cells were able to convert these molecules into H2S. The authors of the study suggested the major beneficial effects of garlic intake, specifically on cardiovascular disease and more broadly on overall health, are mediated by the biological production of H2S.32
A 2007 review described some of the significant physiological actions of H2S.31 It relaxes vascular smooth muscle, induces vasodilation of isolated blood vessels and lowers blood pressure. H2S is also a potent anti-inflammatory and antioxidant molecule that can increase antioxidant defences. Unlike NO, it does not form a potentially harmful toxic metabolite at the low levels generated in tissue. In addition, H2S inhibits apoptosis in a number of cell types and promotes the formation of new blood vessels. Several models of cardiovascular disease have demonstrated significant benefit after the administration of H2S donors. The discovery of this new and novel mechanism of action for garlic adds significant weight to its role in the modification and prevention of cardiovascular disease.
A number of case reports have reflected these effects of garlic on bleeding parameters. A spontaneous spinal epidural haematoma associated with platelet dysfunction from excessive garlic ingestion was reported.33 A patient taking garlic prior to cosmetic surgery experienced bleeding complications and had a clotting time of 12.5 minutes. After cessation of garlic, her clotting time dropped to 6 minutes and there were no complications during a second procedure.34
The value of garlic as a prevention and treatment for cardiovascular diseases will best be determined by controlled clinical trials using cardiovascular morbidity or mortality as endpoints. In the meantime, garlic can be prescribed on the basis that it does favourably influence haemorheological parameters (blood flow characteristics) and some cardiovascular risk factors, including modest effect on serum cholesterol and blood pressure. Attention should be paid to the type of garlic preparation used; the strongest published evidence to date is for garlic powder preparations, although other preparations will also be of value. Caution should be exercised when prescribing garlic to patients who are also taking other blood-thinning medications such as aspirin or warfarin and garlic intake should be discontinued 10 days before surgery. However, a clinical trial with healthy volunteers found no adverse effect for garlic powder (enteric-coated standardised tablets, equivalent 4 g/day of fresh garlic) taken with warfarin.35
When Szent-Gyorgy in the 1930s identified the flavonoid constituents of citrus fruits as a necessary co-factor with ascorbic acid in the prevention of scurvy, he opened an investigation which has actually increased in intensity in recent years. Interest in the flavonols such as rutin and its aglycone quercetin has been augmented by a growing fascination with other phenolic molecules, the oligomeric procyanidins (OPCs) and the polyphenolics linked to the tannins, all very common constituents in dietary fruit and vegetables as well as in herbal remedies.
Flavonoids, a group of phenolic constituents found widely in plants, including most fruits and vegetables, have been found to possess a number of anti-inflammatory effects, including, especially for rutin and others from the flavonol subgroup, effects on the microvasculature36 (see also the discussion in Chapter 2 under Flavonoids).
Rutin, quercetin-3-rutoside, is a flavonoid glycoside with quercetin as an aglycone and rhamnose and glucose as sugar moieties. It is very widely distributed in the plant kingdom. It is official in many pharmacopoeias and is widely sold as a health supplement, sometimes in association with vitamin C. In experiments it has been shown to increase survival times of rats fed a thrombogenic diet and in other animals to reduce oedema, reduce cholesterol-induced atheroma and inhibit the carcinogenic action of benzo(α)pyrene.37 Like ascorbic acid, it is an oxygen radical scavenger and has been shown to reduce the mutagenicity of dusts and asbestos38 and other stressors.39,40
Commercial products with a similar structure, hydroxyethylrutosides or oxerutins (containing principally tri-O-(beta-hydroxyethyl)rutoside, as well as a mixture of mono-, di- and tetra-O-(beta-hydroxyethyl)rutosides), are marketed for the treatment of chronic venous insufficiency. There are a number of reports demonstrating positive effects on capillary permeability,41–43 on venous insufficiency44 and venous hypertension.45 Other researchers have reported an improvement in oxygen perfusion of tissues surrounding varicose veins.46
The development of synthetic rutosides has followed the finding that natural rutin is poorly absorbed. However, over 95% of all polyphenolic intake passes to the colon and is fermented by the gut microflora into simple phenols. For example, rutin is now known to be rapidly metabolised by bacteria in the intestine, via quercetin, to 3,4-dihydroxyphenylacetic acid, a small phenol with antioxidant properties. Simple phenolic acids derived from cinnamic acid (such as gallic acid, salicylic acid, caffeic acid, vanillic acid and ferulic acid, as their esters including chlorogenic acid and rosmarinic acid), with their derived polyphenols, make significantly larger contributions to dietary phenol, polyphenol and tannin intake than the flavonols and flavones upon which the vast majority of attention has been focused. It is important to include these in assessments of the total effects of polyphenols.47 Such degradation products are readily absorbed and are found in urine of animals.48 Early doubts about the venous efficacy of such flavonoid molecules49 have therefore not been sustained. (See also the Pharmacokinetics section of Chapter 2.)
There is much in vitro evidence on the effects of flavonoids and other polyphenolics on the microvasculature that can be noted, but which for reasons above need confirmation clinically. For example:
• hawthorn extract50 and black currant extract51 have NO-mediated vasodilatory effects on rat arteries
• flavonoids inhibit TNF-alpha induced upregulation of the endothelial adhesion mediator ICAM-152
• phloretin from apples reduces endothelial adhesion molecules and platelet activation.53
Compared with the effects of polyphenols in vitro, the effects in vivo are more limited. Several studies, however, show benefits of products in streptozotocin-induced diabetic rats, for example flavonoid-rich citrus fruit extract54 and rooibos tea (Aspalathus linearis).55 A fundamental point is that in vivo studies are not long enough and rarely consider bioavailability problems. In human studies particularly it is important that studies are long term, to more closely reflect the likely effects of dietary consumption of polyphenols. Two critical and important reviews of the intervention studies for the use of polyphenols have been published56,57 (see also the relevant section in Chapter 2).
Short-term observations still point to, if not confirm, prospects of vascular changes. In a prospective, placebo-controlled, randomised study, a high rutoside-containing proprietary product (O-(beta-hydroxyethyl rutosides) at 2 g/day for 6 months was tested on patients with diabetic microangiopathy and oedema. Significant decreases in resting flux and rate of ankle swelling were observed in the active treatment groups.58 In this author’s preliminary observations in 1993, 37 patients suffering symptoms of venous insufficiency entered a clinical study to determine the impact on their microcirculation of buckwheat leaf, a popular natural treatment for this condition that contains high levels of plant flavonoids. Using the mild provocation of a suction cup applied to the lower leg and monitoring changes in local circulatory activity with laser Doppler flowmetry, it was possible to identify three characteristics of vascular responses that differentiated sufferers of venous insufficiency from healthy controls: reduced vascular reactivity, flow resolution rate and vasomotor activity. After establishing baseline levels for these characteristics all the subjects took buckwheat leaf for a total of 6 weeks. Twenty-four satisfactorily completed all stages of the study. In this uncontrolled sample it was possible to demonstrate statistically significant changes in vasomotor activity and vascular reactivity. However, flow resolution rate, that might have indicated a beneficial effect on the endothelium, was not changed.
Both short- and long-term improvements in endothelial function have been seen with doses of green tea catechins equivalent to several cups a day.59 A standardised OPC extract of the bark of the French maritime pine (Pinus pinaster) is known to increase capillary resistance. It has been investigated in five clinical trials with a total number of 1289 patients since the late 1960s for treatment and prevention of diabetic retinal microangiopathy, characterised by vascular lesions with exudate deposits and haemorrhages. All of these studies showed that the extract slowed progression of retinopathy and partly recovered visual acuity. It was shown to improve capillary resistance and reduce leakage into the retina. Tolerance was generally very good and side effects were rare, mostly referring to gastric discomfort.60
Epidemiological studies suggest that the intake of flavonols and flavones is inversely associated with subsequent coronary heart disease (CHD).61 There is, however, mixed evidence for a benefit of increased fruit and vegetable intake in vascular health. Improved endothelial function scores and reduced insulin resistance have been observed in a controlled observation of the effects of the Mediterranean diet over 2 years.62 However, by contrast there was little association found between high fruit and vegetable consumption and the incidence of peripheral arterial disease in a 12-year study of a cohort of 44 059 men initially free of cardiovascular disease and diabetes (after adjustment for smoking and other traditional cardiovascular disease risk factors).63
Overall, it seems most likely that any benefits of polyphenolic intake will follow long-term and relatively substantial use. However, the prospect for short-term changes in vascular reactivity with relatively high doses of polyphenols needs to be further explored.
The above research highlights that therapeutic herbs can offer value in one aspect of the cardiovascular system that is somewhat neglected in modern drug treatment, namely the health of the small blood vessels or microvasculature. As noted above, flavonoids in general, and specifically OPCs from pine bark or grape seed, possess clinically relevant vasoprotective activity. Other plant agents shown to support the microvasculature with, for example, beneficial clinical effects in microangiopathy include bilberry, gotu kola, Ginkgo and garlic (allicin-releasing preparations). They will have application where structures comprising fine blood vessels are affected, such as the retina (e.g. diabetic retinopathy), neurons (e.g. diabetic neuropathy) and the glomeruli of the kidneys (e.g. diabetic nephropathy).
The original observations by William Withering of the benefits of foxglove in the treatment of dropsy by a country herbalist led to the discovery of the digitalis glycosides that became the primary drug treatment for congestive heart failure. Given its seriousness and the potency of these plant extractives, it has generally been accepted that crude herbal drugs no longer have a place in the rational treatment of the condition.
Nevertheless, there is a consistent tradition for the use of herbs with cardiac glycosides such as Convallaria majalis (lily of the valley) around the world and pharmacological cases have been made for their use as broader spectrum remedies (see Chapter 2). Indeed, the use of crude Digitalis folium was favoured by some doctors in Britain over the synthetic isolate until relatively recently. There is also evidence that a wider range of plants may have supportive benefits in the condition. For example, Terminalia arjuna 1500 mg/day demonstrated substantial benefits in the treatment of refractory congestive heart failure linked to dilated cardiomyopathy in a placebo-controlled, double blind crossover trial.64
There is now substantial clinical evidence for the supportive role of hawthorn (Crataegus species) in congestive heart failure, not to supplant conventional medication, but to provide an extra dimension of treatment aimed at supporting the heart muscle itself. (This has been fully reviewed in the hawthorn monograph.)
Other herbs of potential value include Coleus forskohlii containing forskolin, a phytochemical with cardiotonic activity,65 and Salvia miltiorrhiza (dan shen).66 Astragalus also possesses mild cardiotonic activity and can be combined with Korean ginseng for this effect (see the ginseng and Astragalus monographs). Clinical trials from China suggest a clinical benefit from ginseng in patients with congestive heart failure. Mild diuretic herbs may be beneficial for fluid retention, such as dandelion leaves.
Case history
A male patient aged 82 with a history of congestive heart failure, stable angina and poor memory (prescribed aspirin and diuretics) was placed on the following formulation:
Ginkgo biloba (standardised extract) | 2:1 | 40 mL |
Salvia miltiorrhiza | 1:2 | 30 mL |
Panax ginseng | 1:2 | 15 mL |
Astragalus membranaceus | 1:2 | 25 mL |
total | 110 mL |
In addition, hawthorn leaf and flower extract tablets (containing the equivalent of 1 g of herb) were prescribed at two tablets twice daily.
Over a period of 5 years of this treatment (with some variations) the patient has considerably improved, despite his advancing years. His capacity to exercise is now much greater and he recently repainted his timber house. Relatives of the patient have expressed surprise at his marked improvement.
In about 90% of cases with hypertension there is no identifiable cause and the term ‘essential hypertension’ is used. In the remaining cases a cause can be identified and this is known as ‘secondary hypertension’. The main cause is kidney disease; other causes include coarctation of the aorta, endocrine diseases and pregnancy. Generally, the treatment for secondary hypertension is the same as for essential hypertension, but the cause should also be treated if possible. It is important to ensure that patients presenting with essential hypertension do not have a secondary cause. Many patients with hypertension have coexisting cardiovascular risk factors, which should also be addressed.67
Although the milder stages of essential hypertension should probably not be considered as a disease, people with hypertension are more likely than those with normal blood pressure to develop a number of cardiovascular diseases. In particular, hypertension is a risk factor for the development of CHD. As such, it is desirable to treat even mild hypertension.
Treatment should aim for gradual reduction in blood pressure. The kidneys will have become adapted to the previously high levels (indeed, an approach to understanding essential hypertension is that it may be a mechanism to ensure adequate kidney function when this is failing). Sudden reduction could lead to other problems. In this sense natural approaches, if effective, can be doubly suitable.
It is apparent to most practitioners that hypertension is an indication for a broad therapeutic strategy, including dietary and lifestyle advice. Some of the features of this advice are therefore outlined below. It is not advisable to attempt to treat severe (greater than 170/110), malignant or accelerated hypertension with only natural approaches; synthetic prescription drugs can be necessary to avoid serious harm in such cases.
Although the physiological mechanisms responsible for the lowering of blood pressure as a result of exercise are still under debate, strong epidemiological and experimental evidence supports a link between the two.68 Aerobic exercise that uses large muscle groups for 20 to 60 minutes a day for a minimum of 3 days a week is advisable, although there may have to be a gradual build-up to these levels and all stages should be closely monitored.
Obesity and hypertension are strongly linked. There is a continuous linear relationship between excess body fat, blood pressure and the prevalence of hypertension.69 A cause-and-effect relationship has also been demonstrated. Hence weight loss should always be attempted. The waist:hip ratio is a more accurate predictor of hypertension than either body weight or body mass index.70 In extreme cases this is now recognised as linked to the condition known as metabolic syndrome.
The role of sodium (salt) restriction in treating hypertension has been controversial, with recent evidence that casts doubt on the conventional view that low sodium intake is always helpful.71 However, the consensus is that salt reduction does lower high blood pressure readings.72
Randomised, controlled trials indicate a specific blood pressure lowering effect of lactovegetarian diets.73 A non-vegetarian diet rich in fruit and vegetables and low fat dairy products also significantly reduced blood pressure.74 Although the effects of caffeine on blood pressure are considered to be temporary, many clinicians suggest a reduction in caffeine intake to reduce aggravating factors.75 Potassium supplementation or the use of a high-potassium, high-magnesium salt has been shown to reduce blood pressure.76 Increased calcium intake may also be of value77 and 6 g/day of fish oil had a mild lowering effect.78
Relaxation techniques could be valuable, although their acceptance has been hampered by poorly designed and ambiguous studies.79 Some self-prescribed non-prescription drugs may cause or exacerbate hypertension. These include ephedrine, pseudoephedrine and other decongestant and weight loss agents.80
Most of the herbal treatments for hypertension probably act as peripheral vasodilators. They are all slow to exert their activity, except perhaps for Coleus. Important herbs for this condition include the following:
• Crataegus (hawthorn) – as well as reducing high blood pressure this herb has a trophic effect on the heart muscle. This is important because left ventricular heart failure is often caused by prolonged hypertension. The leaves are apparently more potent than the berries for reducing blood pressure (see the hawthorn monograph) and effects are modest.
• Allium sativum (garlic) – as well as its mild antihypertensive effects (see previous) this plant also favourably influences other cardiovascular risk factors. Allicin-releasing preparations are most proven in blood pressure management (see previous discussion).
• Coleus forskohlii – can have a pronounced lowering effect on high blood pressure. Only varieties containing forskolin should be used. Coleus also has pronounced antiplatelet activity, which may be desirable in some cases.65
• Valeriana (valerian) – whether this herb acts as a peripheral or central vasodilator or if the activity is due to a general calming effect on the nervous system is not known. It is usually prescribed for stressed patients (see valerian monograph).
• Olea europaea (olive leaves) – has been proven to lower high blood pressure in clinical trials, provided the dose is sufficiently high.81
• Viburnum opulus (cramp bark) – this herb is thought to relax smooth muscle and has been used to augment antihypertensive prescriptions as a vasorelaxant.
• Achillea millefolium (yarrow) – is used by some herbalists to specifically lower an elevated diastolic blood pressure.
• Taraxacum officinale (dandelion leaves) – has diuretic activity and high levels of potassium and can be useful especially for the treatment of elevated systolic pressure in the elderly.
Other herbs also commonly used to lower high blood pressure include Tilia species (lime flowers) and Viscum album (mistletoe). The Ayurvedic herb Rauwolfia is a powerful treatment for hypertension, but is usually limited to prescription only.
Case history
A female patient aged 48 sought assistance for palpitations, anxiety, angina and mild hypertension. Her ECG did not reveal the presence of a cardiac arrhythmia and her palpitations were less severe in recent times. On examination her blood pressure was 170/95 despite her use of the prescribed drugs labetalol and felodipine.
After treatment over a few months, the following prescription was settled upon:
Ginkgo biloba (standardised extract) | 2:1 | 20 mL |
Panax notoginseng | 1:2 | 20 mL |
Crataegus folia | 1:2 | 25 mL |
Corydalis ambigua | 1:2 | 20 mL |
Hypericum perforatum | 1:2 | 25 mL |
Passiflora incarnata | 1:2 | 20 mL |
Salvia miltiorrhiza | 1:2 | 20 mL |
total | 150 mL |
Dose: 7.5 mL with water three times a day.
Over the ensuing months her blood pressure was typically 135/85. She had no problems with palpitations and her anxiety and angina had improved.
Case history
A male patient aged 62 with blood pressure as high as 160/100 sought herbal treatment instead of the ACE inhibitor offered by his doctor. He had a high stress job as a property developer and also suffered from benign prostatic hyperplasia.
He was prescribed the following:
Crataeva nurvala | 1:2 | 20 mL |
Crataegus folia | 1:2 | 35 mL |
Valeriana officinalis | 1:2 | 20 mL |
Urtica radix | 1:2 | 15 mL |
Zizyphus spinosa | 1:2 | 20 mL |
total | 110 mL |
Dose: 8 mL with water twice per day, combined with separate capsules for his prostate containing Serenoa repens.
After 6 months on the treatment his prostate symptoms were considerably reduced and his average blood pressure was 121/76. The patient also followed the lifestyle and dietary advice outlined above.