Skip to main content
Sage Choice logoLink to Sage Choice
. 2021 Jan 12;41(5):561–581. doi: 10.1177/0333102420970904

Use of medicinal plants for headache, and their potential implication in medication-overuse headache: Evidence from a population-based study in Nepal

Elise Øien Sørnes 1,, Ajay Risal 2,3, Kedar Manandhar 2,3,, Hallie Thomas 1, Timothy J Steiner 1,4, Mattias Linde 1
PMCID: PMC8047708  PMID: 33435708

Abstract

Background

In Nepal, traditional treatment using medicinal plants is popular. Whereas medication-overuse headache is, by definition, caused by excessive use of acute headache medication, we hypothesized that medicinal plants, being pharmacologically active, were as likely a cause.

Methods

We used data from a cross-sectional, nationwide population-based study, which enquired into headache and use of medicinal plants and allopathic medications. We searched the literature for pharmacodynamic actions of the medicinal plants.

Results

Of 2100 participants, 1794 (85.4%) reported headache in the preceding year; 161 (7.7%) reported headache on ≥15 days/month, of whom 28 (17.4%) had used medicinal plants and 117 (72.7%) allopathic medication(s). Of 46 with probable medication-overuse headache, 87.0% (40/46) were using allopathic medication(s) and 13.0% (6/46) medicinal plants, a ratio of 6.7:1, higher than the overall ratio among those with headache of 4.9:1 (912/185). Of 60 plant species identified, 49 were pharmacodynamically active on the central nervous system, with various effects of likely relevance in medication-overuse headache causation.

Conclusions

MPs are potentially a cause of medication-overuse headache, and not to be seen as innocent in this regard. Numbers presumptively affected in Nepal are low but not negligible. This pioneering project provides a starting point for further research to provide needed guidance on use of medicinal plants for headache.

Keywords: Herbal medications, pharmacodynamic activity, overuse, South-East Asia region, Global Campaign against Headache

Background

It has been assumed that medication-overuse headache (MOH) is less of a problem in highly rural poor countries because of the lack of access to pharmaceuticals (1). However, in a large epidemiological study in Nepal, the prevalence of probable MOH (pMOH, defined as the association of headache on ≥15 days/month with overuse of acute medication) was higher than reported in Europe (2,3).

In such countries, there is often strong reliance on alternative and complementary practitioners, and the use of plants for medicinal purposes is common (4,5). If these medicinal plants (MPs) have properties that make them active against acute headache, might their overuse also generate MOH? Studies to answer this are lacking, but the question is of interest also in the Western world, where the use of MPs is increasing (6), with a widespread belief that they are “harmless remedies without side effects” (7). At the same time, MOH is the type of headache associated with the highest cost per person in Europe (8).

In a pioneering project, we used data from a large population-based survey from Nepal to test the hypothesis: “Medicinal plants, being pharmacologically active, are as likely as other medications to cause MOH”. Our public-health objectives were:

  • to establish the proportion of Nepalese adults using MPs against headache

  • to assess the associations of such usage with demographic factors and disease attributes (symptom burden)

  • to identify the MPs used

  • to make a systematic listing of their pharmacological effects on the CNS

  • to ascertain whether and to what extent they might be implicated in MOH causation

Methods

Design of the epidemiological study

This methodology has been described in detail previously (9). In summary, in a cross-sectional, population-based survey, trained health workers visited 2210 households selected randomly from 15 representative districts out of 75 in Nepal (2,9,10). One adult was randomly selected from each household, with 2100 agreeing to participate in a structured interview. The participation proportion was > 99% (2). All data were collected during May 2013.

Instruments

The structured Headache-Attributed Restriction, Disability, Social Handicap and Impaired Participation (HARDSHIP) questionnaire was culturally adapted and translated into Nepali (9). HARDSHIP included demographic factors (age, gender, household consumption and altitude and urbanicity of dwelling) as well as indices of symptom burden (headache frequency, duration and intensity) and use of allopathic medication(s). Queries concerning MPs were added (2): Participants were asked whether they had used any herbal therapies specifically to relieve headache (not taken regularly to prevent headache), their names, if so, and on how many days they had been used during the preceding month.

Diagnosis

We specifically identified participants who reported headache on ≥15 days/month, and, among these, any who were using medications and/or MPs for acute headache. We diagnosed overuse in those who reported: a) Use on ≥15 days/month of either one type of MP or simple analgesics only; or b) use on ≥10 days/month of (i) more than one type of MP; (ii) any other acute medication (such as opioids, ergots or triptans); (iii) a combination of analgesics in different classes or of analgesics with other medications; or (iv) a combination of MPs and analgesics (2).

Migraine or tension-type headache (TTH) were diagnosed according to ICHD-3 beta criteria (11) in those with headache on < 15 days/month. Further information on the methodology has been published elsewhere (2).

Plant identification

Two Nepalese botanists living in Norway were consulted to correct any local plant names that were misspelled in the data and to identify the genus and species of each plant from its local name(s) and description. Synonyms were searched for at www.efloras.org (12).

Literature search

We searched Micromedex and PubMed for reports of any pharmacological effects of the MPs on the CNS. Each plant’s botanical name was used as a search term, with the Medical Subject Heading (MeSH) term “central nervous system” added in the PubMed search. When there were fewer than three matches with the additional search term, only the plant name was used. In cases where we only had the plant-genus, “species” was added as a third search term in cases where there were >150 matches. We also looked for the plants in the encyclopaedia Plants and people of Nepal (13). When the plant names identified by the botanists gave no or few (<3) results, synonyms were searched for. We also looked for potential information sources in the reference lists of relevant articles.

We separated the potential effects of the plants on the CNS into (a) antioxidative effects, (b) anti-inflammatory or immunological effects (anti-inflammatory, pro-inflammatory, immunosuppressive, immunomodulatory effects, or influences on nitric oxide/inducible nitric oxide synthase), (c) effects on receptors, transmitters or synapses in the CNS, (d) effects on vasculature (constrictor or dilator), and (e) other effects.

Data management and statistical analyses

The outcome variables “use of medicinal plant” and “use of allopathic medications” during the preceding month for treatment of headache were categorised as “yes” or “no” and presented as numbers and percentages. Users of both MPs and allopathic medications were included in both categories. Proportions of participants using MPs or allopathic medications, with 95% confidence intervals (CIs), were estimated for any headache, migraine and headache on ≥15 days/month. The independent variable “age” was categorized into five groups (18–25, 26–35, 36–45, 46–55 and 56–65 years). Household consumption per year in United States dollars (USD) (at the time of study: 1 USD ≈ 100 Nepalese rupees) was used as an indicator of participants’ economic status, and categorized into three groups: Poorest (<950 USD/year), poor (950–1200 USD/year), and intermediate and above (>1200 USD/year). This gave the best split in numbers, while taking some account of Nepal’s absolute poverty line (nationally, USD 225 for an individual in 2013, but varying between locations; in Kathmandu, it was USD471 (14)). Our measure of consumption was based on households rather than individuals, and therefore set at a higher level. Dwelling was categorized as urban or rural, and altitude as low (<1000 m) or high (≥1000 m).

The symptom burden of headache was measured by frequency (headache days/month [d/m]), attack duration (hours) and headache intensity. While the first two of these were collected as continuous data, we categorized them so as to yield the best split in numbers between the categories. Thus frequency was categorized into four groups for any headache (<1, 1–2, 3–14 and ≥15 d/m), three for migraine (<1, 1–2 and 3–14 d/m) and two for headache on ≥15 d/m (15–20 and > 20 d/m). Duration was categorized into three groups: For any headache and migraine as < 4, 4–12 and > 12 hours, and for headache on ≥15 d/m as < 48, 48–144 and > 144 hours. We calculated proportion of time in ictal state (pTIS; %) from frequency and mean headache duration, and categorized it, again to give the best split in numbers, into four groups (<1, 1–3, 3.1–10 and > 10%) for any headache and migraine, and three groups (<60, 60–80 and > 80%) for headache on ≥15 days/month. Intensity was categorized as “not bad”, “quite bad” and “very bad” (equating to “mild”, “moderate” and “severe”).

We used bivariate and multivariate logistic regression analyses (with odds ratios [OR] and adjusted odds ratios [AOR], each with 95% CIs) to investigate the associations of plant or medication use on the one hand, in all headache, migraine and headache on ≥15 days/month, and demographic variables and indices of headache burden on the other. These variables were entered as covariates in the multivariate logistic regression analyses, although, for frequency and duration, pTIS was excluded, and, for pTIS, frequency and duration were excluded.

In view of the number of analyses, we considered p < 0.02 to be statistically significant. Two-tailed p-values were calculated. All data were analysed with SPSS 21.0 software (IBM Corp, Armonk, NY, USA).

Results

Participants

A total of 2100 adults aged 18–65 years were included in the nationwide study; their sociodemographic characteristics have been presented previously (206). Here are analyzed data of 1794 participants (85.4%) who reported headache during the preceding year. Their mean age was 36.1 ± 12.6 years, and male/female ratio was 1:1.6.

Use and overuse of medicinal plants and allopathic medications

Of these 1794 participants, 185 (10.3%) also reported use of MP(s) for headache at least once in the preceding month, while 912 (50.8%) had used allopathic medication(s) (Table 1). Each was more likely to be used by those with headache types expected to be more troublesome. Thus, of the 728 with migraine, 85 (11.7%) had used MP(s) and 423 (58.1%) had used allopathic medication(s) (Table 2). Of the 161 participants reporting headache on ≥15 days/month, 28 (17.4%) had used MP(s) and 117 (72.7%) had used allopathic medication(s) (Table 3). We did not separately analyse those with TTH.

Table 1.

Use of medicinal plants and allopathic medication at least once in the preceding month among participants with any headache according to demographic factors and symptom burden (N = 1794).

Variable N
Medicinal plants

Allopathic medication
n (%)
Bivariate analyses

Ethnobotany in theNepal Himalaya.Multivariate analyses
n (%)
Bivariate analyses

Multivariate analyses
OR (95% CI) p AOR* (95% CI) p OR (95% CI) p AOR* (95% CI) p
Yes 1794 185 (10.3) 912 (50.8)
Age (in years)
 18–25 426 28 (6.6) Reference Reference 205 (48.1) Reference Reference
 26–35 581 58 (10.0) 1.6 (0.9–2.5) 0.057 1.6 (0.9–2.6) 0.055 307 (52.8) 1.2 (0.9–1.6) 0.14 1.2 (0.9–1.6) 0.20
 36–45 358 46 (12.8) 2.1 (1.3–3.4) 0.003 2.0 (1.2–3.6) 0.006 173 (48.3) 1.0 (0.8–1.3) 0.96 0.9 (0.7–1.3) 0.58
 46–55 258 30 (11.6) 1.9 (1.1–3.2) 0.023 1.6 (0.9–2.9) 0.078 138 (53.5) 1.2 (0.9–1.7) 0.17 1.2 (0.8–1.7) 0.34
 56–65 171 23 (13.5) 2.2 (1.2–3.9) 0.008 1.9 (1.1–3.5) 0.037 89 (52.0) 1.2 (0.8–1.7) 0.34 1.0 (0.7–1.4) 0.91
Gender
 Male 689 82 (11.9) Reference Reference 306 (44.4) Reference Reference
 Female 1105 103 (9.3) 0.8 (0.6–1.1) 0.081 0.8 (0.6-1.1) 0.11 606 (54.8) 1.5 (1.31.8) < 0.001 1.3 (1.1–1.6) 0.014
Household consumption (USD/year)
 950–1200 687 70 (10.2) Reference Reference 347 (50.5) Reference Reference
 <950 686 76 (11.1) 1.1 (0.8–1.5) 0.60 0.9 (0.6–1.3) 0.67 355 (51.7) 1.1 (0.9–1.3) 0.65 1.0 (0.8–1.3) 0.97
 >1200 421 39 (9.3) 0.9 (0.6–1.4) 0.62 0.9 (0.6–1.4) 0.70 210 (49.9) 1.0 (0.8–1.2) 0.85 1.1 (0.8–1.4) 0.71
Dwelling
 Urban 686 36 (5.2) Reference Reference 299 (43.6) Reference Reference
 Rural 1108 149 (13.4) 2.8 (1.9–4.1) <0.001 2.6 (1.8–3.9) <0.001 613 (55.3) 1.6 (1.3–1.9) <0.001 1.7 (1.4–2.1) <0.001
Household altitude
 <1000 m 845 63 (7.5) Reference Reference 393 (46.5) Reference Reference
 ≥1000 m 949 122 (12.9) 1.8 (1.3–2.5) <0.001 1.5 (1.1–2.2) 0.015 519 (54.7) 1.4 (1.2–1.7) 0.001 1.1 (0.9–1.4) 0.34
Headache burden indices
Frequency (days/month)
 <1 626 46 (7.3) Reference Reference 203 (32.4) Reference Reference
 1–2 531 61 (11.5) 1.6 (1.1–2.4) 0.016 1.5 (0.9–2.4) 0.082 285 (53.7) 2.4 (1.9–3.1) <0.001 1.8 (1.3–2.3) <0.001
 3–14 478 50 (10.5) 1.5 (0.9–2.3) 0.067 1.2 (0.7–2.0) 0.49 307 (64.5) 3.8 (2.9–4.9) <0.001 2.1 (1.6–2.9) <0.001
 ≥15 161 28 (17.4) 2.7 (1.6–4.4) <0.001 2.0 (1.1–3.8) 0.015 117 (72.7) 5.5 (3.8–8.1) <0.001 2.3 (1.5–3.7) <0.001
Duration (in hours)
 <4 590 43 (7.3) Reference Reference 183 (31.0) Reference Reference
 4–12 466 47 (10.1) 1.4 (0.9–2.2) 0.11 1.2 (0.7–2.1) 0.50 227 (48.7) 2.1 (1.6–2.7) <0.001 1.6 (1.2–2.1) 0.001
 >12 738 95 (12.9) 1.9 (1.3–2.7) 0.001 2.2 (0.9–5.4) 0.080 502 (68.0) 4.7 (3.7–6.0) <0.001 2.5 (1.8–3.4) <0.001
Intensity
 Not bad 373 32 (8.6) Reference Reference 130 (34.9) Reference Reference
 Quite bad 901 83 (9.2) 1.1 (0.7–1.7) 0.72 0.9 (0.6–1.4) 0.68 451 (50.1) 1.9 (1.5–2.4) <0.001 1.3 (1.1–1.7) 0.047
 Very bad 520 70 (13.5) 1.7 (1.1–2.6) 0.025 1.1 (0.7–1.8) 0.76 331 (63.7) 3.3 (2.5–4.3) <0.001 1.7 (1.3–2.4) 0.001
Proportion (%) of time in ictal state
 <1 785 58 (7.4) Reference Reference 264 (33.6) Reference Reference
 1–3 219 27 (12.3) 1.8 (1.1–2.9) 0.022 1.8 (1.1–3.0) 0.021 118 (53.9) 2.3 (1.7–3.1) <0.001 2.2 (1.6–3.0) <0.001
 3.1–10 403 49 (12.2) 1.7 (1.2–2.6) 0.007 1.7 (1.1–2.7) 0.013 252 (62.5) 3.3 (2.6–4.2) <0.001 3.0 (2.3–3.9) <0.001
 >10 387 51 (13.2) 1.9 (1.3–2.8) 0.002 2.0 (1.3–30) 0.002 278 (71.8) 5.0 (3.3–6.6) <0.001 4.4 (3.3–5.9) <0.001

AOR: adjusted odds ratio: OR: odds ratio; CI: confidence interval. N = total number within subsample; n = number within subsample responding positively.

*

Adjusted for age, gender, household consumption, dwelling, altitude, headache frequency (F), attack duration (D), intensity and proportion of time in ictal state (pTIS) (for F and D, pTIS was excluded; for pTIS, F and D were excluded). p-values are two-tailed, and emboldened when significant (<0.02).

Table 2.

Use of medicinal plants and allopathic medication at least once in the preceding month among participants with migraine according to demographic factors and symptom burden (n = 728).

Variable N
Medicinal plants

Allopathic medication
n (%)
Bivariate analyses

Multivariate analyses
n (%)
Bivariate analyses

Multivariate analyses
OR (95% CI) p AOR* (95% CI) p OR (95% CI) p AOR* (95% CI) p
Yes 728 85 (11.7) 423 (58.1)
Age (in years)
 18–25 153 9 (5.9) Reference Reference 88 (57.5) Reference Reference
 26–35 241 29 (12.0) 2.2 (1.1–4.8) 0.048 2.1 (1.0–4.8) 0.063 143 (59.3) 1.1(0.7–1.6) 0.72 1.1 (0.7–1.8) 0.57
 36–45 158 22 (13.9) 2.6 (1.2–5.8) 0.021 2.8 (1.2–6.6) 0.014 87 (55.1) 0.9 (0.6–1.4) 0.66 0.9 (0.6–1.5) 0.73
 46–55 101 16 (15.8) 3.0 (1.3–7.1) 0.012 2.8 (1.2–6.8) 0.024 64 (63.4) 1.3 (0.8–2.1) 0.35 1.4 (0.8–2.4) 0.26
 56–65 75 9 (12.0) 2.1(0.8–5.7) 0.11 2.0 (0.7–5.5) 0.18 41 (54.7) 0.9 (0.5–1.6) 0.68 0.6 (0.4–1.2) 0.15
Gender
 Male 249 35 (14.1) Reference Reference 141 (56.6) Reference Reference
 Female 479 50 (10.4) 0.7 (0.5–1.1) 0.15 0.8 (0.5–1.3) 0.36 282 (58.9) 1.1 (0.8–1.5) 0.56 1.0 (0.7–1.4) 0.99
Household consumption (USD/year)
 950–1200 277 32 (11.6) Reference Reference 166 (59.9) Reference Reference
 <950 289 33 (11.4) 1.0 (0.6–1.7) 0.96 0.8 (0.5–1.4) 0.38 169 (58.5) 1.3 (0.9–1.9) 0.25 0.8 (0.6–1.2) 0.37
 >1200 162 20 (12.3) 1.1 (0.6–2.0) 0.80 1.2 (0.6–2.2) 0.63 88 (54.3) 1.2 (0.8–1.8) 0.39 0.8 (0.5–1.2) 0.21
Dwelling
 Urban 261 13 (5.0) Reference Reference 135 (51.7) Reference Reference
 Rural 467 72 (15.4) 3.5 (1.9–6.4) <0.001 3.4 (1.8–6.5) <0.001 288 (61.7) 1.5 (1.1–2.0) 0.009 1.5 (1.1-2.1) 0.021
Household altitude
 <1000 m 287 21 (7.3) Reference Reference 145 (50.5) Reference Reference
 ≥1000 m 441 64 (14.5) 2.2 (1.3–3.6) 0.004 1.9 (1.1–3.2) 0.027 278 (63.0) 1.7 (1.2–2.3) 0.001 1.3 (0.9–1.9) 0.085
Headache burden indices
Frequency (days/month)
 <1 235 23 (9.8) Reference Reference 92 (39.1) Reference Reference
 1–2 226 31 (13.7) 1.5 (0.8–2.6) 0.19 1.2 (0.6–2.5) 0.62 140 (61.9) 2.5 (1.7–3.7) <0.001 2.3 (1.5–3.6) <0.001
 3–14 267 31 (11.6) 1.2 (0.7–2.1) 0.51 0.8 (0.4–1.8) 0.58 191 (71.5) 3.9 (2.7–5.7) <0.001 3.2 (1.9–5.2) <0.001
Duration (in hours)
 <4 148 12 (8.1) Reference Reference 61 (41.2) Reference Reference
 4–12 225 26 (11.6) 1.5 (0.7–3.0) 0.28 1.5 (0.7–3.2) 0.35 118 (52.4) 1.6 (1.1–2.4) 0.034 1.2 (0.7–1.8) 0.53
 >12 355 47 (13.2) 1.7 (0.9–3.4) 0.11 1.8 (0.7–4.3) 0.20 244 (68.7) 3.1 (2.1–4.7) <0.001 1.4 (0.8–2.4) 0.23
Intensity
 Not bad 60 4 (6.7) Reference Reference 27 (45.0) Reference Reference
 Quite bad 354 40 (11.3) 1.8 (0.6–5.2) 0.29 1.5 (0.5–4.5) 0.47 195 (55.1) 1.5 (0.9–2.6) 0.15 1.3 (0.7–2.3) 0.40
 Very bad 314 41 (13.1) 2.1 (0.7–6.1) 0.17 1.5 (0.5–4.6) 0.46 201 (64.0) 2.2 (1.2–3.8) 0.006 1.7 (0.9–3.1) 0.094
Proportion (%) of time in ictal state
 <1 263 24 (9.1) Reference Reference 111 (42.2) Reference Reference
 1–3 83 11 (13.3) 1.5 (0.7–3.3) 0.28 1.5 (0.7–3.2) 0.34 50 (60.2) 2.1 (1.3–3.4) 0.004 2.2 (1.3–3.7) 0.003
 3.1–10 248 36 (14.5) 1.7 (1.0–2.9) 0.061 1.6 (0.9–2.9) 0.12 161 (64.9) 2.5 (1.8–3.6) <0.001 2.4 (1.7–3.5) <0.001
 >10 134 14 (10.4) 1.2 (0.6–2.3) 0.67 1.1 (0.6–2.4) 0.72 101 (75.4) 4.2 (2.6–6.6) <0.001 4.4 (2.7–7.1) <0.001

AOR: adjusted odds ratio; OR: odds ratio; CI: confidence interval. N = total number within subsample; n = number within subsample responding positively.

*

Adjusted for age, gender, household consumption, dwelling, altitude, headache frequency (F), attack duration (D), intensity and proportion of time in ictal state (pTIS) (for F and D, pTIS was excluded; for pTIS, F and D were excluded). p-values are two-tailed, and emboldened when significant (<0.02).

Table 3.

Use of medicinal plants and allopathic medication at least once in the preceding month among participants with headache on ≥ 15 days/month according to demographic factors and symptom burden (n = 161).

Variable N
Medicinal plants

Allopathic medication
n (%)
Bivariate analyses

Multivariate analyses
n (%)
Bivariate analyses

Multivariate analyses
OR (95% CI) p AOR* (95% CI) p OR (95 % CI) p AOR* (95% CI) p
Yes 161 28 (17.4) 117 (72.7)
Age (in years)
 18–25 29 3 (10.3) Reference Reference 17 (58.6) Reference Reference
 26–35 51 10 (19.6) 2.1 (0.5–8.4) 0.29 2.4 (0.6–10.6) 0.23 38 (74.5) 2.1 (0.8–5.4) 0.14 1.5 (0.5–4.6) 0.47
 36–45 34 9 (26.5) 3.1 (0.8–12.9) 0.12 2.6 (0.6–12.0) 0.21 24 (70.6) 1.7 (0.6–4.8) 0.32 1.4 (0.4–4.5) 0.58
 46–55 29 5 (17.2) 1.8 (0.4–8.3) 0.45 2.4 (0.5–12.6) 0.29 22 (75.9) 2.2 (0.7–6.8) 0.17 1.6 (0.5–5.6) 0.47
 56–65 18 1 (5.6) 0.5 (0.1–5.3) 0.57 0.6 (0.1–6.7) 0.67 16 (88.9) 5.6 (1.1–29.3) 0.039 7.8 (1.2–50.6) 0.029
Gender
 Male 44 10 (22.7) Reference Reference 26 (59.1) Reference Reference
 Female 117 18 (15.4) 0.6 (0.3–1.5) 0.28 0.5 (0.2–1.6) 0.25 91 (77.8) 2.4 (1.2–5.1) 0.019 1.8 (0.7–4.7) 0.21
Household consumption (USD/year)
 950–1200 59 10 (16.9) Reference Reference 41 (69.5) Reference Reference
 <950 59 13 (22.0) 1.4 (0.9–2.2) 0.11 1.2 (0.7–2.1) 0.50 41 (69.5) 1.0 (0.5–2.2) 1.0 1.2 (0.5–2.9) 0.75
 >1200 43 5 (11.6) 1.9 (1.3–2.7) 0.001 2.2 (0.9–5.4) 0.080 35 (81.4) 1.9 (0.7–4.9) 0.18 1.9 (0.6–5.8) 0.25
Dwelling
 Urban 54 5 (9.3) Reference Reference 42 (77.8) Reference Reference
 Rural 107 23 (21.5) 2.7 (0.9–7.5) 0.060 2.1 (0.7–6.5) 0.20 75 (70.1) 0.7 (0.3–1.4) 0.18 0.7 (0.3–1.7) 0.39
Household altitude
 <1000 m 69 12 (17.4) Reference Reference 50 (72.5) Reference Reference
 ≥1000 m 92 16 (17.4) 1.0 (0.4–2.3) 1.0 0.9 (0.3-2.2) 0.74 67 (72.8) 1.1 (0.5-2.1) 0.30 1.2 (0.5–2.6) 0.72
Headache burden indices
Frequency (days/month)
 15–20 99 24 (24.2) Reference Reference 73 (73.7) Reference Reference
 >20 62 4 (6.5) 0.2 (0.1–0.7) 0.007 0.4 (0.1–1.6) 0.18 44 (71.0) 0.9 (0.4–1.8) 0.70 0.2 (0.1–0.7) 0.010
Duration (in hours)
 <48 56 9 (16.1) Reference Reference 31 (55.4) Reference Reference
 48–144 50 15 (30.0) 2.2 (0.9–5.7) 0.091 2.6 (0.9–7.5) 0.089 40 (80.0) 3.2 (1.4–7.7) 0.008 2.6 (1.0–6.9) 0.053
 >144 55 4 (7.3) 0.4 (0.1–1.4) 0.16 0.9 (0.2–4.6) 0.91 46 (83.6) 4.1 (1.7–10.1) 0.002 8.6 (2.1–34.7) 0.003
Intensity
 Not bad/quite bad 62 9 (14.5) Reference Reference 41 (66.1) Reference Reference
 Very bad 99 19 (19.2) 1.4 (0.6–3.3) 0.45 1.5 (0.6–4.0) 0.43 76 (76.8) 1.7 (0.8–3.4) 0.14 1.3 (0.6–3.1) 0.49
Proportion (%) of time in ictal state
 <60 64 14 (21.9) Reference Reference 46 (71.9) Reference Reference
 60–80 45 11 (24.4) 1.2 (0.5–2.8) 0.75 1.3 (0.5–3.4) 0.61 30 (66.7) 0.8 (0.3–1.8) 0.56 0.7 (0.3–1.7) 0.68
 >80 52 3 (5.8) 0.2 (0.1–0.8) 0.023 0.2 (0.1–0.9) 0.041 41 (78.8) 1.5 (0.6–3.5) 0.39 0.8 (0.3–2.2) 0.71

AOR: adjusted odds ratio: OR: odds ratio; CI: confidence interval. N = total number within subsample; n = number within subsample responding positively.

*

Adjusted for age, gender, household consumption, dwelling, altitude, headache frequency (F), attack duration (D), intensity and proportion of time in ictal state (pTIS) (for F and D, pTIS was excluded; for pTIS, F and D were excluded). p-values are two-tailed, and emboldened when significant (<0.02).

Of the 117 participants with headache on ≥15 days/month and using allopathic medication(s), 40 (34.2%) met our criteria for overuse. Of the 28 with headache on ≥15 days/month and using MP(s), six (21.4%) met our criteria for overuse. Three of these also reported use of allopathic medication(s), one on 15 days/month and therefore meeting our criteria for overuse of these also.

Plant identification

From the 73 local plant names reported, the botanists were able to identify or partially identify 45 plants (61.6%) and unable to identify 28. Of the 45, five were identified only by their genera, three were given only probable identities, and one was only partly identified (Herbal products Vicks). The identities of two others ‒ “banphool” and “ketaki” ‒ were not confirmed by the botanists but, in the views of the two Nepalese authors (AR and KM), very likely nonetheless, and accepted since these were relevant to the hypothesis; therefore, 47 local plant names (64.4%) in total were at least partially identified.

Four of the local plant names were applied to mixtures of different species, two were identified with more than one probable botanical name, two were used for more than one species and two for the same species as two others. Zandu balm was an exception, since this was found to contain both exact species and a mix of different species in a genus. Taken together, there were 60 species (four of them uncertain because of uncertain identifications by the botanists and six without definite confirmation by the botanists) and five genera identified from the 47 local plant names.

Literature search

We reviewed 191 publications or encyclopaedias in the literature search. Of the 60 species, 49 were identified pharmacodynamically. Of the five genera, only two were identified pharmacodynamically since there were few studies on the genera as a whole. We found only one randomized controlled trial (RCT) relevant to use for headache, which was of low quality. No meta-analyses were found. The remainder of the studies found were animal, in vitro, or case studies (Table 4).

Table 4.

Medicinal plants used for headache, with their botanical names (local names in parenthesis), pharmacodynamic properties and reported adverse events.

Formulation Used for headache (source and level of evidence (15)) Assumed pharmacodynamic mechanisms on CNS Reported adverse events on CNS (serious: SAEs [including death]; non-serious: NSAEs)
Abelmoschus moschatus Medik* (component of Navaratna oil)
Topical (13) No data Antioxidative (16)
Abrus precatorius L. (Rato gerdi)
Oral/topical (13) (Level V) (15) Immunostimulating (17); receptors (17) SAEs: Death (1820); coma (18,19); seizures (18)  NSAEs: Fever, reduced mental status (18)
Aconitum ferox Wall. Ex. Ser. (Bikma herbal)
Oral/topical (13,18,21) No data Other (18,2226); Receptors (27,28) SAEs: Death, paralysis (18); muscular fasciculations, tonic-clonic seizures (18,25)  NSAEs: Paresthesias, pain, severe headache, restlessness, apprehension, confusion, incoordination, miosis, mydriasis, diplopia, blurred vision, yellow-green vision (18)
Acorus calamus L. (Bojo)
Oral/parenteral/topical (13,18) (Level V) (18) Other (29,30) SAEs: None reported (18)  NSAEs: None reported (18)
Allium sativum L.*** (Garlic)
Oral/topical (13,18) (Level V) (31) Antioxidative (18,3235); anti-inflammatory (3638); receptors (33,35); other (31,39); antiapoptotic (35,36) SAEs: None reported on CNS (18)  NSAEs: Headache, fatigue, vertigo (18)
Allium wallichii Kunth*** (Garlic)
Oral (13) No data Antioxidative (40) No data
Aloe vera L. Burm. f. (Aloe vera herbal)
Oral/topical (13,18,41) No data Antioxidative (4245); anti-inflammatory (18,43,46); other (42,47) SAEs: none reported (18,41)  NSAEs: none reported (18,41)
Artemisia indica Willd. (Titepati leaves)
Oral/topical (13) (Level V) (48) Antioxidative (49); immunostimulating (50); receptors (51) No data
Azadirachta indica A. Juss. (Neem leaves)
Oral/topical (13,18) (Level V) (13) Antioxidative (52); anti-inflammatory (53,54); receptors (55); antiapoptotic (52,53) SAEs: Death, Reye-like syndrome, altered consciousness, seizures, decreased responsiveness (18)  NSAEs: Lethargy (18)
Brassica napus L. (Mustard paste)
No data No data No data No data
Calotropis gigantea L. Dryand (Aank)
Inhaled/oral/topical (13) No data Receptors (56) No data
Centella asiatica L. Urb. (Gortapre)
Oral/parenteral/topical (13,18) (Level V) (13) Antioxidative (5761); anti-inflammatory (18,62); receptors (6366); antiapoptotic (62) No data
Cheilanthes albomarginata C.B. Clarke (Rani sini herbal)
Oral/topical (13) No data Antioxidative (67); anti-inflammatory (67) No data
Colebrookea oppositifolia Sm. (Dursur)
Intranasal/oral/parenteral/topical (13) (Level V) (13) Receptors (68) No data
Curcuma longa L. (Turmeric herbal)
Oral/Parenteral (18) No data Antioxidative (18,6972); anti-inflammatory (18,6971,7377); receptors (7881); other (69,8285); vascular (77) SAEs: None reported (18,41,86)  NSAEs: None reported (5,41,86)
Curcuma zedoaria (Christm.) Roscoe* (component of Navaratna oil)
No data No data Antioxidative (87) No data
Cymbopogon pendulus (Nees ex Steud.) W. Watson (Lemon grass)
No data No data No data No data
Cyperus rotundus L.* (component of Navaratna oil)
Oral (13) No data Antioxidative (88,89); anti-inflammatory (89,90); receptors (91,92); other (93); antiapoptotic (89) No data
Eclipta prostrata L.* (component of Navaratna oil)
Oral/topical (13) No data Antioxidative (94); receptors (94); other (95) No data
Elettaria cardamomum L. (Aalainch)
Oral (18,41) No data Antioxidative (18,96); anti-inflammatory (97,98); other (99) SAEs: None reported (18)  NSAEs: None reported (18)
Eucalyptus globulus Labill.* (component of Zandu balm, from “Herbal products Vick”)
Oral/parenteral/topical (18,41) No data Antioxidative (18); anti-inflammatory (18); receptors (100) SAEs: Loss of consciousness, hypoventilation, convulsions (101,102)  NSAEs: Ataxia, CNS depression (101)
Gaultheria fragrantissima Wall.* (component of Zandu balm, from “Herbal products Vick”)
Topical (48) (Level V) (48) Antioxidative (103,104) No data
Helianthus annuus L. (Sunflower)
No data No data Antioxidative (105); anti-inflammatory (105) No data
Hibiscus rosa-sinensis L.* (component of Navaratna oil)
Oral/topical (13) No data Antioxidative (106); receptors (107) No data
Hordeum vulgare L.* (Jamara)
Oral/topical (18) No data Antioxidative (18); other (108) SAEs: None reported (18)  NSAEs: None reported (18)
Inula cappa (Buch.-Ham. ex D.Don) DC. (Dwareko jaro)
Oral/topical (13) (Level V) (2) Anti-inflammatory (109,110) No data
Lilium polyphyllum D. Don* (component of Navaratna oil)
No data No data No data No data
Lygodium japonicum (Thumb.) Sw.** (Pinase)
Topical (13) No data Anti-inflammatory (111) No data
Lysimachia alternifolia Wall.** (Pinase)
Inhaled (13) No data No data No data
Mentha arvensis L.* (component of Navaratna oil)
Intranasal/oral/topical (18,41) No data Antioxidative (112,113); anti-inflammatory (113,114); receptors (113,115,116); other (114) SAEs: None reported (41)  NSAEs: None reported (41)
Mentha spicata L. (Menthol oil)
Oral (13) No data Oxidative (110) No data
Micromeria biflora (Buch.-Ham. Ex. D. Don) Benth.** (Pinase)
Inhaled/oral/topical (13) No data No data No data
Myrica esculenta Buch.-Ham. Ex. D. Don (Kafal ko bokra)
Oral/topical (13,18) (Level V) (13) Antioxidative (117); anti-inflammatory (118) No data
(Neo)picrorhiza scrophulariiflora (Pennel) D.Y. Hong (Kurki)
Oral/topical (13) No data No data No data
Nyctanthes arbor-tristis L. (Parijaat leaves)
Oral/topical (13) No data Antioxidative (119); immunostimulating (120122); anti-inflammatory (123); other (124) No data
Ocimum tenuiflorum L. (Tulsi leaves/Kapoor*, ingredient in Banphool oil [translation not approved by botanists])
Oral/topical (13,48) (Level V) (48) Antioxidative (125); anti-inflammatory (18); receptors (126129) No data
Ophiocordyceps sinensis (Berk.) G.H. Sung, J.M. Sung, Hywel-Jones & Spatafora (mushroom) (Yarsagumba)
No data No data Antioxidative (130,131); anti-inflammatory (132); immunostimulating (132); other (132) No data
Parmelia perlata* (Huds.) Ach. (component of Navaratna oil)
No data No data No data No data
Phyllanthus emblica L.* (component of Navaratna oil/Aamla, which is also a component of Banphool oil), and in monotherapy
Oral (13) No data Antioxidative (133137); anti-inflammatory (135137); receptors (138); other (135,137,138) No data
Plumbago zeylanica L. (Chitu)
Oral/topical (13) No data Anti-inflammatory (139,140); receptors (141) No data
Rubus ellipticus Sm. (Ainselu root)
Oral/topical (13,18) No data Antioxidative (142,143) No data
Swertia angustifolia Buch.-Ham. Ex D. Don** (Chiraito)
Oral (13) No data No data No data
Swertia chirayita Buch.-Ham. ex D. Don** (Chiraito)
Oral/topical (13) (Level V) (13), (Level V) (144) Antioxidative (145); anti-inflammatory (146,147) SAEs: None reported (144)  NSAEs: No data
Syzygium nervosum A Cunn, ex DC. (Kyamuna ko munto)
No data No data Antioxidative (148) No data
Terminalia chebula Retz. (Harro)
Oral/topical (13) No data Antioxidative (149,150); other (150) No data
Triticum aestivum L.* (Jamara)
Topical (18) No data Receptors (151) SAEs: No data NSAEs: Transformation of episodic into daily headache (152)
Vitex negundo L. (Simali)
Inhaled/oral/topical (13) (Level V) (13) Antioxidative (153,154); anti-inflammatory (154,155); receptors (155) No data
Zanthoxylum armatum DC. (Timur)
Oral/topical (13) No data Antioxidative (156,157); anti-inflammatory (158,159) No data
Zea mays L.* (Jamara)
No data No data Antioxidative, antiapoptotic (160) No data
Zingiber officinale Roscoe (Ginger)
Oral/topical (13,41) (level V) (18); (level IIb) (161) (RCT) Antioxidative (162,163); anti-inflammatory (162,164168); receptors (169171); other (172) SAEs: None on CNS (161) (RCT)  NSAEs: None on CNS (161) (RCT)
Genera with uncertain translations
Ageratum sp. (Bherapate herbal)
No data No data No data No data
Cotoneaster sp. (Ghareko jaro or Ghare herbal)
No data No data No data No data
Hedyotis sp. (Nimaniko jadhibuti)
No data No data No data No data
Mentha sp. L.* (component of Zandu balm from “Herbal products Vick”/Satva Pudina*; component of Banphool oil [translation not approved by botanists])
No data No data Anti-inflammatory (173); receptors (173) SAEs: Coma (18)  NSAEs: Ataxia, confusion, vertigo, CNS depression (18)
Piper sp. (Pepper)
No data No data Anti-inflammatory (174)
Species with uncertain translations
Asparagus racemosus Willd.*** (Satawari [participants called it Sartawa])
Oral (13,18) No data Antioxidative (175,176); receptors (176179) SAEs: None reported (18)  NSAEs: None reported (18)
Ipomoea carnea Jacq. (Ajamari jhar [participants called it Ajawane])
Topical (13) No data Antioxidative (180); anti-inflammatory (180); other (180) No data.
Justicia adhatoda L. (Asurako or Asuro)
Oral/topical (13) No data No data No data
Paris polyphylla Sm.***(Satuwa [participants called it Sartawa])
Oral/topical (13) No data No data No data
Translations not approved by the botanists
Convolvulus pluricaulis Choisy* (181) (component of Banphool oil (182))
No data No data Antioxidative (183,184); receptors (185,186); other (184,187190) No data
Nardostachys grandiflora D.C.* (181) (component of Banphool oil (182))
No data No data No data No data
Pandanus fascicularis Lam. (181) (Ketaki (191))
No data No data Antiinflammatory (192) No data
Santalum album L.* (181) (component of Banphool oil (182))
No data No data Other (193) SAEs: CNS depression, seizures, coma (18)  NSAEs: None reported (18)
Syzygium aromaticum L. Merrill & Perry* (component of Banphool oil (182))
Oral/topical (18) (Level V) (208) Antioxidative (194,195); anti-inflammatory (18); receptors (196); other (197,198) SAEs: Coma, seizures (199) NSAEs: Slight CNS depression (200)
Terminalia bellirica (Gaertn.) Roxb.* (181) (Barro (181), component of Banphool oil (182))
No data No data Antioxidative (201,202); anti-inflammatory (203,204); receptors (205) No data
Formulation Used for headache (level of evidence (15)) Assumed pharmacodynamic mechanisms on CNS Reported adverse events (AEs) on CNS, or death

NSAEs: non-serious adverse events; RCT: randomised controlled trial; SAEs: serious adverse events.

*

Not in monotherapy.

**

Two or more different species have same local name.

***

More than one possible botanical name.

Of the 60 species, 37 (61.7%) were shown in the literature to have antioxidative effects, 27 (45.0%) to have anti-inflammatory or immunosuppressive effects, and 23 (38.3%) to have effects on receptors, transmitters or synapses in the CNS (Table 4). Of the five genera, two were shown to have anti-inflammatory or immunosuppressive effects and one to have effects on receptors, transmitters or synapses in the CNS (Table 4). Fifteen species (25.0%) were mentioned in the literature as used against headache (Table 4). Serious adverse events (SAEs) affecting the CNS or resulting in death were reported for seven species (coma, loss of consciousness, seizures, paralyses, death) (Table 4). In addition, Allium sativum L. (garlic) has been shown to have anticoagulant properties resulting in bleeding (18). Respiratory and cardiac arrest have also been reported after ingestion of Azadirachta indica A. Juss (18), as has disseminated intravascular coagulopathy after ingestion of Syzygium aromaticum (L) Merrill & Perry (199).

Systemic bioavailability is implied by these pharmacodynamic properties. This is a key issue. We did not collect data on routes of administration to avoid overloading the already-long enquiry. While the literature indicates that many preparations of these MPs would be applied topically, others are taken orally and some are prepared for inhalation. Table 4 lists only the routes of administration potentially relevant to MOH causation.

Associations according to headache type

Any headache

We found somewhat greater use of allopathic medications (AOR 1.3), but not of MPs, among females than males (Table 1). There were no clear associations with age or household consumption.

Higher proportions of participants with any headache used MPs (AOR 2.6) and allopathic medications (AOR 1.7) in rural areas than urban (Table 1). Associations with high altitude, apparent in bivariate analysis, lost significance in multivariate analysis.

Increasing headache frequency (from 1 to ≥15 days/month) was clearly associated with increasingly probable use of allopathic medication(s) (from 32.4% to 72.7%, a factor of 2.2; AOR 1.8–2.3), but, for MPs, this was so only for frequency ≥15 days/month (from 7.3% to 17.4%, a factor of 2.4; AOR 2.0; Table 1). Long-duration (AOR 2.5) and severe headache (AOR 1.7) were each associated with increased use of allopathic medication(s), but not (in multivariate analyses) of MPs (Table 1). Participants with pTIS ≥1% were more likely to have used allopathic medication(s) (AOR 2.2–4.4); a similar trend for MPs became clearly significant only when pTIS exceeded 3% (AOR 1.7–2.0; Table 1).

Migraine

There were no clear associations with age, gender or household consumption.

A higher proportion of participants with migraine used MPs (AOR 3.4) in rural areas than urban, but for allopathic medications this association just lost significance in multivariate analysis (AOR 1.5; p = 0.021; Table 2). Associations with high altitude, apparent in bivariate analysis, once more lost significance in multivariate analysis. High frequency (AOR 3.2) and pTIS ≥1% were associated with more probable use of allopathic medications (AOR 2.2–4.4) but not of MPs (Table 2).

Headache on ≥15 days/month

Only one association was clear: Less use of allopathic medication(s) (AOR 0.2) in very high-frequency headache (> 20 days/month) (Table 3). A similar trend for MPs (AOR 0.2) did not survive multivariate analysis. A trend away from use of MPs (AOR 0.2) with very high pTIS (>80%) was discernible but did not meet our significance threshold (p > 0.02; Table 3).

Are medicinal plants as likely as other medications to be associated with headache on ≥15 days/month?

As noted earlier, of 912 participants with headache and using allopathic medication(s), 117 (12.8%) reported headache on ≥15 days/month with 40 of these (34.2%) meeting our criteria for overuse; of 185 using MP(s), 28 (15.1%) reported headache on ≥15 days/month, six (21.4%) meeting our criteria for overuse. The difference between these proportions was not significant (p = 0.259 [chi-squared]). Of the 46 with headache on ≥15 days/month and overuse, 87.0% (40/46) were using allopathic medication(s) and 13.0% (6/46) were using MP(s), a ratio of 6.7:1, somewhat higher than the overall usage ratio of 4.9:1 (912/185) (again not significant: p = 0.262).

Discussion

The study found that a considerable minority (10.3%) of people with headache in Nepal had used MPs as treatment for it during the preceding month, although a much higher proportion (50.8%) had used allopathic medication(s). As might be expected, use of each was more likely (MPs: 17.4%; allopathic medication(s) 72.7%) among those with headache on ≥15 days/month. Among those with migraine in particular, use of MPs was positively associated with both rural (AOR 3.4) and high-altitude (AOR 1.9) dwelling.

Use of MPs for headache is therefore common, although less so than use of allopathic medications. Use of MPs is especially common among those with headache on ≥15 days/month, again less so than allopathic medications. Both MPs and allopathic medications were overused, according to our definitions, in association with headache on ≥15 days/month.

However, answers to the questions of whether MPs might be implicated in MOH causation, raised by our fifth objective, and, if so, whether they are as likely to cause MOH as allopathic medications, raised in our starting hypothesis, depend on evidence not only of use, overuse and association but also of a potential causal mechanism. We note, before further consideration of these, that diagnosis of MOH is presumptive, even with allopathic medications: the diagnostic criteria of ICHD-3 beta for practical reasons omit evidence of causation, relying on “Not better accounted for by another ICHD-3 diagnosis” (11). In epidemiology, this last criterion cannot be reliably applied, hence the term “probable MOH”.

With this caveat, we address these questions.

Of 60 plant species identified, 49 were pharmacodynamically active on the CNS, with effects (on CNS receptors, and anti-inflammatory and antioxidative actions) likely to be relevant in MOH causation. This is a revelation of some importance, since 8.8% of all adults aged 18–65 years in Nepal (10.3% of the 85.4% with headache) apparently use MPs for headache (although only 25.0% of these plants have been recognized in the literature as treatments used for headache, and, in a study of traditional use of MPs in Western Nepal, headache was not listed as an indication for any of the plants (207)).

The probably multiple mechanisms of MOH causation are still unclear. Every known drug used in acute headache treatment, with a range of pharmacological actions, can cause MOH when overused (208,209). The MPs used for headache by our study participants are highly pharmacologically active, many with actions similar to those of conventional medications for headache: Inter alia, anti-inflammatory, serotonergic and opioidergic properties, also seen in headache medications such as aspirin, NSAIDs, triptans and opioids. While we cannot (and do not) conclude that MPs are an actual cause of MOH, these findings are such that MPs, wherever they are used and overused, must be included among its potential causes.

It is worth observing that, while only six participants using MPs had headache on ≥15 days/month and fulfilled our criteria for overuse, there is no accepted definition of overuse of MPs. We placed MPs on equal terms with other acute headache medication (2), so that use of MPs of a single type on ≥15 days/month or of more than one type on ≥10 days/month was deemed to be overuse. This said, our search of the literature indicated that many of the plants used for headache had multiple, independent pharmacological actions. This made it likely that each consisted of different active substances, and more logical, therefore, to use ≥10 days/month as the overuse threshold regardless of how many types were consumed, increasing the number diagnosed as MP-overusers. However, we considered the conservative definition a better test of our hypothesis.

Use of MPs for headache escalated with headache frequency at the same rate as use of allopathic medication(s), more than doubling as frequency increased from > 1 to ≥15 days/month. As, probably, the most influential driver of medication overuse, increasing headache frequency appears to operate similarly on MPs and allopathic medications. However, people in ictal state for most of the time (pTIS > 80%) had, apparently, given up on MPs. This was not seen with allopathic medications. It is easy to speculate that people in this near-end state, with headache almost all of their time, find (or believe they find) greater benefit from allopathic medications, so that many switch allegiance. Since there was no sign of reduced use of MPs as pTIS increased up to 80%, there is no support for the alternative explanation – that risk of developing more frequent headache is higher in those who use allopathic medication(s) than in those using MPs. There are no other studies on MPs and headache to throw light on this question.

Other associations were, perhaps, of limited interest. Considerably higher proportions of participants with migraine used MPs in rural areas than urban (AOR 3.5), a finding that was not unexpected, and a reflection, probably, of urban/rural cultural difference driven, in part, by less-easy access in rural areas to health care. Association with high altitude lost significance in multivariate analysis because rural dwelling was itself strongly associated with higher altitude.

There were limitations in this study. First, even though there were 2100 participants overall, the sample size of those reporting headache on ≥15 days per month was only 161. This resulted in lower than ideal statistical power. A second limitation lay in language and plant identification. Since no data collected in the original survey related the local names of the MPs to the ethnicity of those using them, it was impossible for the botanists to identify all of them with their botanical names (more than 120 languages are spoken as mother tongue in Nepal (210)). In addition, no data had been collected on dosages or routes of administration, as already noted, or on whether the MPs were gathered by the participants themselves, prescribed by health workers or dispensed by traditional healers. Although this was information relevant to our public-health objectives, the original survey had deferred its collection to future studies (if warranted) to avoid over-loading an already lengthy enquiry.

A third factor, also limiting but outside our control, was the lack of guiding evidence in the literature. Only one relevant RCT was found in the search for side effects and use against headache, and this was of low quality. It was impossible to establish whether the MPs are actually beneficial in acute headache. Additionally, there were no studies on the pharmacological effects on the CNS for a large proportion of the MPs, which left uncertainties about their possible actions.

Despite these limitations, a participation proportion of 99.6% in the original survey excluded participation bias (2), and the methodology for collecting the data was thoroughly considered (211). These were considerable strengths. This is the first large nationwide study to investigate the use of MPs for headache in Nepal. We assume it will be the starting point for multiple endeavours: Further studies on overuse of plants with pharmacological properties among people with headache disorders; RCTs on MPs used for headache to establish their effects, if any; basic research on the mechanisms that lead to MOH to test our hypothesis more decisively, and provision of guidance on the use and misuse of MPs for headache.

Conclusion

MPs are used as symptomatic treatments for headache by almost 9% of the Nepalese population aged 18–65 years, 15% of whom report headache on ≥15 days/month. Many of these MPs have pharmacodynamic properties similar to those implicated in MOH causation by allopathic medication overuse. On this accumulation of evidence, MPs, whenever used, should be considered at least a potential cause of MOH, perhaps no less than allopathic medications, although this is unproven. The six presumptive cases we identified in Nepal, in a sample of 2100 (0.3%), are, in public-health terms, a small but not negligible proportion. Given a global context in which herbal remedies are becoming increasingly popular, this evidence attaches considerable importance to further research on MOH in relation to MPs. This study provides a starting point, where none existed before.

Public health relevance

  • Medicinal plants must be included among the potential causes of MOH.

  • In Nepal, where use of medicinal plants is culturally entrenched, presumptive evidence suggests a small but nonetheless measurable impact at population level.

  • In developed countries, where MOH is the headache with the highest cost per person, the use of medicinal plants is widely believed to be harmless, and increasing.

  • For these reasons, further research and guidance on use of MPs for headache are needed.

Abbreviations

AE: adverse events; AOR: adjusted odds ratio; CI: confidence interval; CNS: central nervous system; D: duration; F: headache frequency (headache days); HARDSHIP: Headache-Attributed Restriction, Disability, Social Handicap and Impaired Participation; IRC-KUSMS: Institutional Review Committee of Kathmandu University School of Medical Sciences; MeSH: Medical Subject Heading; MOH: medication-overuse headache; MP: medicinal plant; NSAEs: non-serious adverse events; OR: odds ratio; pMOH: probable MOH; pTIS: proportion of time in ictal state; RCT: randomized controlled trial; SAEs: serious adverse events; TTH: tension-type headache; USD: United States dollar.

Acknowledgements

This study was conducted in collaboration with the Global Campaign against Headache, led by the non-governmental organization Lifting The Burden in official relations with the World Health Organization and with Dhulikhel Hospital and Kathmandu University Hospital, Nepal.

We are grateful to Jan Ove Rein, Senior Research Librarian, Sindre Andre Pedersen, Senior Research Librarian and Katrine Aronsen, Head Librarian (all at The Medicine and Health Library, NTNU) for their practical assistance and help with the literature search, as well as the botanists, Kamal Prasad Acharya and Dilli Prasad Rijal (UiT – The Arctic University of Norway), for their great assistance in identifying plants from their local names, and for valuable advice.

Footnotes

Author contributions: All authors were involved in conception and design of this study. KM, AR, ML and TJS were responsible for data acquisition in the original population-based study. EØS planned and conducted the literature search and established the pharmacological properties of medicinal plants reportedly used by participants. KM and HT performed the analyses. All authors contributed to interpretation of the data. EØS drafted the manuscript and TJS contributed to its revision. All authors reviewed it critically for intellectual content and all, other than KM, approved the final version of the manuscript.

Declaration of conflicting interests: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: TJS is a Director and Trustee of Lifting The Burden. The authors declare no other competing interests.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This project was financially supported by Samarbeidsorganet, the Liaison Committee between the Central Norway Regional Health Authority and the Norwegian University of Science and Technology (NTNU) (grant no. 653010-46060911).

Availability of data and materials: The datasets used and analysed during this study are held securely at The Norwegian University of Science and Technology. They are still undergoing analyses. Once these are completed, the datasets will be available from the corresponding author for academic purposes.

Ethics and approvals: The study protocol was approved by the Regional Committee for Health and Research Ethics in Central Norway, the Nepal Health Research Council and the Institutional Review Committee of Kathmandu University School of Medical Sciences, Dhulikhel Hospital (IRC-KUSMS). Consents were taken according to requirements of IRC-KUSMS (2,10).

ORCID iD: Elise Øien Sørnes https://orcid.org/0000-0003-0803-5745

References

  • 1.Jones JM. Headaches beyond the roads. Headache 2010; 50: 1219–1220. [DOI] [PubMed] [Google Scholar]
  • 2.Manandhar K, Risal A, Steiner TJ, et al. The prevalence of primary headache disorders in Nepal: A nationwide population-based study. J Headache Pain 2015; 16: 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Stovner LJ, Andree C. Prevalence of headache in Europe: A review for the Eurolight project. J Headache Pain 2010; 11: 289–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sharma UR, Malla KJ, Uprety RK. Conservation and management efforts of medicinal and aromatic plants in Nepal. Banko Janakari 2017; 14; 3–11. [Google Scholar]
  • 5.Kunwar RM, Bussmann RW. Ethnobotany in the Nepal Himalaya. J Ethnobiol Ethnomed 2008; 4: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ekor M. The growing use of herbal medicines: issues relating to adverse reactions and challenges in monitoring safety. Front Pharmacol 2014; 4: 177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Furst R, Zundorf I. Evidence-based phytotherapy in Europe: Where do we stand? Planta Med 2015; 81: 962–967. [DOI] [PubMed] [Google Scholar]
  • 8.Linde M, Gustavsson A, Stovner LJ, et al. The cost of headache disorders in Europe: The Eurolight project. Eur J Neurol 2012; 19: 703–711. [DOI] [PubMed] [Google Scholar]
  • 9.Manandhar K, Risal A, Steiner TJ, et al. Estimating the prevalence and burden of major disorders of the brain in Nepal: Methodology of a nationwide population-based study. J Headache Pain 2014; 15: 52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Linde M, Edvinsson L, Manandhar K, et al. Migraine associated with altitude: Results from a population-based study in Nepal. Eur J Neurol 2017; 24: 1055–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Headache Classification Committee of the International Headache Society. The International Classification of Headache Disorders, 3rd edition (beta version). Cephalalgia 2013; 33: 629–808. [DOI] [PubMed]
  • 12.Press JR, Shrestha KK, Sutton DA. Annotated checklist of the flowering plants of Nepal, http://www.efloras.org/flora_page.aspx?flora_id=110 (accessed 3 April 2018).
  • 13.Manandhar NP, Manandhar S. Plants and people of Nepal. Portland, OR: Timber Press, 2002, pp.1–599. [Google Scholar]
  • 14.Pokharel T. Poverty in Nepal: Characteristics and challenges. J Poverty Invest Dev 2015; 11. [Google Scholar]
  • 15.Centre for Evidence-Based Medicine. Oxford Centre for Evidence-based Medicine – levels of evidence (March 2009), https://www.cebm.net/2009/06/oxford-centre-evidence-based-medicine-levels-evidence-march-2009/ (2009, accessed 10 June 2018).
  • 16.Gul MZ, Bhakshu LM, Ahmad F, et al. Evaluation of Abelmoschus moschatus extracts for antioxidant, free radical scavenging, antimicrobial and antiproliferative activities using in vitro assays. BMC Complement Altern Med 2011; 11: 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bhasker AS, Sant B, Yadav P, et al. Plant toxin abrin induced oxidative stress mediated neurodegenerative changes in mice. Neurotoxicology 2014; 44: 194–203. [DOI] [PubMed] [Google Scholar]
  • 18.Truven Health Analytics. Micromedex® 2.0, (electronic version), http://www.micromedexsolutions.com/ (2018, accessed 22 February 2018).
  • 19.Sahni V, Agarwal SK, Singh NP, et al. Acute demyelinating encephalitis after jequirity pea ingestion (Abrus precatorius). Clin Toxicol (Phila) 2007; 45: 77–79. [DOI] [PubMed] [Google Scholar]
  • 20.Dickers KJ, Bradberry SM, Rice P, et al. Abrin poisoning. Toxicol Rev 2003; 22: 137–142. [DOI] [PubMed] [Google Scholar]
  • 21.Chan TY. Aconitum alkaloid poisoning related to the culinary uses of aconite roots. Toxins 2014; 6: 2605–2611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fu M, Wu M, Qiao Y, et al. Toxicological mechanisms of Aconitum alkaloids. Die Pharmazie 2006; 61: 735–741. [PubMed] [Google Scholar]
  • 23.Nyirimigabo E, Xu Y, Li Y, et al. A review on phytochemistry, pharmacology and toxicology studies of Aconitum. J Pharm Pharmacol 2015; 67: 1–19. [DOI] [PubMed] [Google Scholar]
  • 24.Yamanaka H, Doi A, Ishibashi H, et al. Aconitine facilitates spontaneous transmitter release at rat ventromedial hypothalamic neurons. Brit J Pharmacol 2002; 135: 816–822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ameri A. The effects of Aconitum alkaloids on the central nervous system. Prog Neurobiol 1998; 56: 211–235. [DOI] [PubMed] [Google Scholar]
  • 26.Suk KD, Yoon KC, Shin JP, et al. Aconite induced myelo-optic neuropathy in a rabbit model. Korean J Ophthalmol 1994; 8: 77–82. [DOI] [PubMed] [Google Scholar]
  • 27.Zhao XY, Wang Y, Li Y, et al. Songorine, a diterpenoid alkaloid of the genus Aconitum, is a novel GABA(A) receptor antagonist in rat brain. Neurosci Lett 2003; 337: 33–36. [DOI] [PubMed] [Google Scholar]
  • 28.Li TF, Fan H, Wang YX. Aconitum-derived Bulleyaconitine A exhibits antihypersensitivity through direct stimulating Dynorphin A expression in spinal microglia. J Pain 2016; 17: 530–548. [DOI] [PubMed] [Google Scholar]
  • 29.Reddy S, Rao G, Shetty B, et al. Effects of Acorus calamus rhizome extract on the neuromodulatory system in restraint stress male rats. Turkish Neurosurg 2015; 25: 425–431. [DOI] [PubMed] [Google Scholar]
  • 30.Hazra R, Ray K, Guha D. Inhibitory role of Acorus calamus in ferric chloride-induced epileptogenesis in rat. Hum Experim Toxicol 2007; 26: 947–953. [DOI] [PubMed] [Google Scholar]
  • 31.Marschollek C, Karimzadeh F, Jafarian M, et al. Effects of garlic extract on spreading depression: In vitro and in vivo investigations. Nutr Neurosci 2017; 20: 127–134. [DOI] [PubMed] [Google Scholar]
  • 32.Pintana H, Sripetchwandee J, Supakul L, et al. Garlic extract attenuates brain mitochondrial dysfunction and cognitive deficit in obese-insulin resistant rats. Appl Physiol Nutr Metab 2014; 39: 1373–1379. [DOI] [PubMed] [Google Scholar]
  • 33.Mukherjee D, Banerjee S. Learning and memory promoting effects of crude garlic extract. Indian J Experim Biol 2013; 51: 1094–1100. [PubMed] [Google Scholar]
  • 34.Brunetti L, Menghini L, Orlando G, et al. Antioxidant effects of garlic in young and aged rat brain in vitro. J Med Food 2009; 12: 1166–1169. [DOI] [PubMed] [Google Scholar]
  • 35.Liu SG, Ren PY, Wang GY, et al. Allicin protects spinal cord neurons from glutamate-induced oxidative stress through regulating the heat shock protein 70/inducible nitric oxide synthase pathway. Food Funct 2015; 6: 321–330. [DOI] [PubMed] [Google Scholar]
  • 36.Zhang B, Li F, Zhao W, et al. Protective effects of allicin against ischemic stroke in a rat model of middle cerebral artery occlusion. Mol Med Rep 2015; 12: 3734–3738. [DOI] [PubMed] [Google Scholar]
  • 37.Nillert N, Pannangrong W, Welbat JU, et al. Neuroprotective effects of aged garlic extract on cognitive dysfunction and neuroinflammation induced by beta-amyloid in rats. Nutrients 2017; 9: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Yun HM, Ban JO, Park KR, et al. Pharmacol Ther 2014; 142: 183–195. [DOI] [PubMed]
  • 39.Thorajak P, Pannangrong W, Welbat JU, et al. Effects of aged garlic extract on cholinergic, glutamatergic and GABAergic systems with regard to cognitive impairment in Abeta-induced rats. Nutrients 2017; 9: 686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Bhandari J, Muhammad B, Thapa P, et al. Study of phytochemical, anti-microbial, anti-oxidant, and anti-cancer properties of Allium wallichii. BMC Complem Alt Med 2017; 17: 102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Blumenthal MB and Werner R. The complete German Commission E monographs. 1st ed. Austin, TX: American Botanical Council Integrative Medicine Communications, 1998, pp.1–685.
  • 42.Rathor N, Arora T, Manocha S, et al. Anticonvulsant activity of Aloe vera leaf extract in acute and chronic models of epilepsy in mice. J Pharm Pharmacol 2014; 66: 477–485. [DOI] [PubMed] [Google Scholar]
  • 43.Yuksel Y, Guven M, Kaymaz B, et al. Effects of Aloe vera on spinal cord ischemia-reperfusion injury of rats. J Invest Surg 2016; 29: 389–398. [DOI] [PubMed] [Google Scholar]
  • 44.Lee KY, Weintraub ST, Yu BP. Isolation and identification of a phenolic antioxidant from Aloe barbadensis. Free Radic Biol Med 2000; 28: 261–265. [DOI] [PubMed] [Google Scholar]
  • 45.Wang Y, Cao L, Du G. Protective effects of Aloe vera extract on mitochondria of neuronal cells and rat brain. Zhongguo Zhong Yao Za Zhi 2010; 35: 364–368. [DOI] [PubMed] [Google Scholar]
  • 46.Mirshafiey A, Aghily B, Namaki S, et al. Therapeutic approach by Aloe vera in experimental model of multiple sclerosis. Immunopharmacol Immunotoxicol 2010; 32: 410–415. [DOI] [PubMed] [Google Scholar]
  • 47.Halder S, Mehta AK, Mediratta PK. Aloe vera improves memory and reduces depression in mice. Nutr Neurosci 2013; 16: 250–254. [DOI] [PubMed] [Google Scholar]
  • 48.The World Conservation Union (IUCN). National register of medicinal plants, https://portals.iucn.org/library/sites/library/files/documents/2000-058.pdf (2000, accessed 11 February 2020).
  • 49.Rashid S, Rather MA, Shah WA, et al. Chemical composition, antimicrobial, cytotoxic and antioxidant activities of the essential oil of Artemisia indica Willd. Food Chem 2013; 138: 693–700. [DOI] [PubMed] [Google Scholar]
  • 50.Ruwali P, Ambwani TK, Gautam P. In vitro immunomodulatory potential of Artemisia indica Willd. in chicken lymphocytes. Vet World 2018; 11: 80–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Khan I, Karim N, Ahmad W, et al. GABA-A receptor modulation and anticonvulsant, anxiolytic, and antidepressant activities of constituents from Artemisia indica Linn. Evid-based Complem Alt Med 2016; 2016: 1215393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Vaibhav K, Shrivastava P, Khan A, et al. Azadirachta indica mitigates behavioral impairments, oxidative damage, histological alterations and apoptosis in focal cerebral ischemia-reperfusion model of rats. Neurol Sci 2013; 34: 1321–1330. [DOI] [PubMed] [Google Scholar]
  • 53.Bedri S, Khalil EA, Khalid SA, et al. Azadirachta indica ethanolic extract protects neurons from apoptosis and mitigates brain swelling in experimental cerebral malaria. Malar J 2013; 12: 298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yanpallewar S, Rai S, Kumar M, et al. Neuroprotective effect of Azadirachta indica on cerebral post-ischemic reperfusion and hypoperfusion in rats. Life Sci 2005; 76: 1325–1338. [DOI] [PubMed] [Google Scholar]
  • 55.Pillai NR, Santhakumari G, Laping J. Some pharmacological actions of ‘nimbidin’ – a bitter principle of Azadirachta indica-a juss (neem). Anc Sci Life 1984; 4: 88–95. [PMC free article] [PubMed] [Google Scholar]
  • 56.Argal A, Pathak AK. CNS activity of Calotropis gigantea roots. J Ethnopharmacol 2006; 106: 142–145. [DOI] [PubMed] [Google Scholar]
  • 57.Doknark S, Mingmalairak S, Vattanajun A, et al. Study of ameliorating effects of ethanolic extract of Centella asiatica on learning and memory deficit in animal models. J Med Assoc Thailand 2014; 97: S68–S76. [PubMed] [Google Scholar]
  • 58.Gray NE, Sampath H, Zweig JA, et al. Centella asiatica attenuates amyloid-beta-induced oxidative stress and mitochondrial dysfunction. J Alzheimer’s Dis 2015; 45: 933–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Dhanasekaran M, Holcomb LA, Hitt AR, et al. Centella asiatica extract selectively decreases amyloid beta levels in hippocampus of Alzheimer’s disease animal model. Phytother Res 2009; 23: 14–19. [DOI] [PubMed] [Google Scholar]
  • 60.Ramanathan M, Sivakumar S, Anandvijayakumar PR, et al. Neuroprotective evaluation of standardized extract of Centella asciatica in monosodium glutamate treated rats. Indian J Experim Biol 2007; 45: 425–431. [PubMed] [Google Scholar]
  • 61.Gray NE, Harris CJ, Quinn JF, et al. Centella asiatica modulates antioxidant and mitochondrial pathways and improves cognitive function in mice. J Ethnopharmacol 2016; 180: 78–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Song D, Jiang X, Liu Y, et al. Asiaticoside attenuates cell growth inhibition and apoptosis induced by Abeta1-42 via inhibiting the TLR4/NF-kappaB signaling pathway in human brain microvascular endothelial cells. Front Pharmacol 2018; 9: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chatterjee TK, Chakraborty A, Pathak M, et al. Effects of plant extract Centella asiatica (Linn.) on cold restraint stress ulcer in rats. Indian J Experim Biol 1992; 30: 889–891. [PubMed] [Google Scholar]
  • 64.Nasir MN, Abdullah J, Habsah M, et al. Inhibitory effect of Asiatic acid on acetylcholinesterase, excitatory post synaptic potential and locomotor activity. Phytomedicine 2012; 19: 311–316. [DOI] [PubMed] [Google Scholar]
  • 65.Visweswari G, Prasad KS, Chetan PS, et al. Evaluation of the anticonvulsant effect of Centella asiatica (gotu kola) in pentylenetetrazol-induced seizures with respect to cholinergic neurotransmission. Epilepsy Behav 2010; 17: 332–335. [DOI] [PubMed] [Google Scholar]
  • 66.Awad R, Levac D, Cybulska P, et al. Effects of traditionally used anxiolytic botanicals on enzymes of the gamma-aminobutyric acid (GABA) system. Can J Physiol Pharmacol 2007; 85: 933–942. [DOI] [PubMed] [Google Scholar]
  • 67.Lamichhane R, Kim SG, Poudel A, et al. Evaluation of in vitro and in vivo biological activities of Cheilanthes albomarginata Clarke. BMC Complement Altern Med 2014; 14: 342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Viswanatha GL, Venkataranganna MV, Prasad NBL. Ameliorative potential of Colebrookea oppositifolia methanolic root extract against experimental models of epilepsy: Possible role of GABA mediated mechanism. Biomed Pharmacother 2017; 90: 455–465. [DOI] [PubMed] [Google Scholar]
  • 69.Mythri RB, Bharath MM. Curcumin: A potential neuroprotective agent in Parkinson’s disease. Curr Pharmaceut Des 2012; 18: 91–99. [DOI] [PubMed] [Google Scholar]
  • 70.Yu S, Wang X, He X, et al. Curcumin exerts anti-inflammatory and antioxidative properties in 1-methyl-4-phenylpyridinium ion (MPP(+))-stimulated mesencephalic astrocytes by interference with TLR4 and downstream signaling pathway. Cell Stress Chap 2016; 21: 697–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Wang XS, Zhang ZR, Zhang MM, et al. Neuroprotective properties of curcumin in toxin-base animal models of Parkinson’s disease: A systematic experiment literatures review. BMC Complement Altern Med 2017; 17: 412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Ma XW, Guo RY. Dose-dependent effect of Curcuma longa for the treatment of Parkinson’s disease. Experim Ther Med 2017; 13: 1799–1805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ammon HPT, Wahl MA. Pharmacology of Curcuma longa. Planta Medica 1991; 57: 1–7. [DOI] [PubMed] [Google Scholar]
  • 74.Witkin JM, Li X. Curcumin, an active constituent of the ancient medicinal herb Curcuma longa L.: Some uses and the establishment and biological basis of medical efficacy. CNS Neurol Dis Drug Targets 2013; 12: 487–497. [DOI] [PubMed] [Google Scholar]
  • 75.Xie L, Li XK, Funeshima-Fuji N, et al. Amelioration of experimental autoimmune encephalomyelitis by curcumin treatment through inhibition of IL-17 production. Int Immunopharmacol 2009; 9: 575–581. [DOI] [PubMed] [Google Scholar]
  • 76.Yu SY, Gao R, Zhang L, et al. Curcumin ameliorates ethanol-induced memory deficits and enhanced brain nitric oxide synthase activity in mice. Prog Neuro-Psychopharmacol Biol Psych 2013; 44: 210–216. [DOI] [PubMed] [Google Scholar]
  • 77.Jiang J, Wang W, Sun YJ, et al. Neuroprotective effect of curcumin on focal cerebral ischemic rats by preventing blood-brain barrier damage. Eur J Pharmacol 2007; 561: 54–62. [DOI] [PubMed] [Google Scholar]
  • 78.Pyrzanowska J, Piechal A, Blecharz-Klin K, et al. The influence of the long-term administration of Curcuma longa extract on learning and spatial memory as well as the concentration of brain neurotransmitters and level of plasma corticosterone in aged rats. Pharmacol Biochem Behav 2010; 95: 351–358. [DOI] [PubMed] [Google Scholar]
  • 79.Yu ZF, Kong LD, Chen Y. Antidepressant activity of aqueous extracts of Curcuma longa in mice. J Ethnopharmacol 2002; 83: 161–165. [DOI] [PubMed] [Google Scholar]
  • 80.Zhao X, Wang C, Zhang JF, et al. Chronic curcumin treatment normalizes depression-like behaviors in mice with mononeuropathy: Involvement of supraspinal serotonergic system and GABAA receptor. Psychopharmacology 2014; 231: 2171–2187. [DOI] [PubMed] [Google Scholar]
  • 81.Lin TY, Lu CW, Wang CC, et al. Curcumin inhibits glutamate release in nerve terminals from rat prefrontal cortex: possible relevance to its antidepressant mechanism. Prog Neuro-Psychopharmacol Biol Psych 2011; 35: 1785–1793. [DOI] [PubMed] [Google Scholar]
  • 82.Nabiuni M, Nazari Z, Safaeinejad Z, et al. Curcumin downregulates aquaporin-1 expression in cultured rat choroid plexus cells. J Med Food 2013; 16: 504–510. [DOI] [PubMed] [Google Scholar]
  • 83.Kim DS, Kim JY, Han Y. Curcuminoids in neurodegenerative diseases. Rec Pat CNS Drug Discov 2012; 7: 184–204. [DOI] [PubMed] [Google Scholar]
  • 84.Nam SM, Choi JH, Yoo DY, et al. Effects of curcumin (Curcuma longa) on learning and spatial memory as well as cell proliferation and neuroblast differentiation in adult and aged mice by upregulating brain-derived neurotrophic factor and CREB signaling. J Med Food 2014; 17: 641–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Li Y, Li J, Li S, et al. Curcumin attenuates glutamate neurotoxicity in the hippocampus by suppression of ER stress-associated TXNIP/NLRP3 inflammasome activation in a manner dependent on AMPK. Toxicol Applied Pharmacol 2015; 286: 53–63. [DOI] [PubMed] [Google Scholar]
  • 86.Goel A, Kunnumakkara AB, Aggarwal BB. Curcumin as “curecumin”: From kitchen to clinic. Biochem Pharmacol 2008; 75: 787–809. [DOI] [PubMed] [Google Scholar]
  • 87.Toda S. Inhibitory effects of aromatic herbs on lipid peroxidation and protein oxidative modification of mice brain homogenate by copper in vitro. Phytother Res 2001; 15: 541–543. [DOI] [PubMed] [Google Scholar]
  • 88.Sunil AG, Kesavanarayanan KS, Kalaivani P, et al. Total oligomeric flavonoids of Cyperus rotundus ameliorates neurological deficits, excitotoxicity and behavioral alterations induced by cerebral ischemic-reperfusion injury in rats. Brain Res Bull 2011; 84: 394–405. [DOI] [PubMed] [Google Scholar]
  • 89.Lee CH, Hwang DS, Kim HG, et al. Protective effect of Cyperi rhizoma against 6-hydroxydopamine-induced neuronal damage. J Med Food 2010; 13: 564–571. [DOI] [PubMed] [Google Scholar]
  • 90.Azimi A, Ghaffari SM, Riazi GH, et al. Alpha-cyperone of Cyperus rotundus is an effective candidate for reduction of inflammation by destabilization of microtubule fibers in brain. J Ethnopharmacol 2016; 194: 219–227. [DOI] [PubMed] [Google Scholar]
  • 91.Ha JH, Lee KY, Choi HC, et al. Modulation of radioligand binding to the GABA(A)-benzodiazepine receptor complex by a new component from Cyperus rotundus. Biol Pharm Bull 2002; 25: 128–130. [DOI] [PubMed] [Google Scholar]
  • 92.Pal D, Dutta S, Sarkar A. Evaluation of CNS activities of ethanol extract of roots and rhizomes of Cyperus rotundus in mice. Acta Pol Pharm 2009; 66: 535–541. [PubMed] [Google Scholar]
  • 93.Ngamrojanavanich N, Manakit S, Pornpakakul S, et al. Inhibitory effects of selected Thai medicinal plants on Na+,K+-ATPase. Fitoterapia 2006; 77: 481–483. [DOI] [PubMed] [Google Scholar]
  • 94.Kim DI, Lee SH, Hong JH, et al. The butanol fraction of Eclipta prostrata (Linn) increases the formation of brain acetylcholine and decreases oxidative stress in the brain and serum of cesarean-derived rats. Nutr Res 2010; 30: 579–584. [DOI] [PubMed] [Google Scholar]
  • 95.Jung WY, Kim H, Park HJ, et al. The ethanolic extract of the Eclipta prostrata L. ameliorates the cognitive impairment in mice induced by scopolamine. J Ethnopharmacol 2016; 190: 165–173. [DOI] [PubMed] [Google Scholar]
  • 96.Verma SK, Jain V, Katewa SS. Blood pressure lowering, fibrinolysis enhancing and antioxidant activities of cardamom (Elettaria cardamomum). Indian J Biochem Biophys 2009; 46: 503–506. [PubMed] [Google Scholar]
  • 97.Majdalawieh AF, Carr RI. In vitro investigation of the potential immunomodulatory and anti-cancer activities of black pepper (Piper nigrum) and cardamom (Elettaria cardamomum). J Med Food 2010; 13: 371–381. [DOI] [PubMed] [Google Scholar]
  • 98.Daoudi A, Aarab L, Abdel-Sattar E. Screening of immunomodulatory activity of total and protein extracts of some Moroccan medicinal plants. Toxicol Ind Health 2013; 29: 245–253. [DOI] [PubMed] [Google Scholar]
  • 99.Masoumi-Ardakani Y, Mahmoudvand H, Mirzaei A, et al. The effect of Elettaria cardamomum extract on anxiety-like behavior in a rat model of post-traumatic stress disorder. Biomed Pharmacother 2017; 87: 489–495. [DOI] [PubMed] [Google Scholar]
  • 100.Girish C, Raj V, Arya J, et al. Evidence for the involvement of the monoaminergic system, but not the opioid system in the antidepressant-like activity of ellagic acid in mice. Eur J Pharmacol 2012; 682: 118–125. [DOI] [PubMed] [Google Scholar]
  • 101.Tibballs J. Clinical effects and management of eucalyptus oil ingestion in infants and young children. Med J Australia 1995; 163: 177–180. [DOI] [PubMed] [Google Scholar]
  • 102.Patel S, Wiggins J. Eucalyptus oil poisoning. Arch Dis Child 1980; 55: 405–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pandey BP, Thapa R, Upreti A. Chemical composition, antioxidant and antibacterial activities of essential oil and methanol extract of Artemisia vulgaris and Gaultheria fragrantissima collected from Nepal. Asian Pacific J Trop Med 2017; 10: 952–959. [DOI] [PubMed] [Google Scholar]
  • 104.Cong F, Joshi KR, Devkota HP, et al. Dhasingreoside: New flavonoid from the stems and leaves of Gaultheria fragrantissima. Nat Prod Res 2015; 29: 1442–1448. [DOI] [PubMed] [Google Scholar]
  • 105.Guo S, Ge Y, Na Jom K. A review of phytochemistry, metabolite changes, and medicinal uses of the common sunflower seed and sprouts (Helianthus annuus L.). Chem Cent J 2017; 11: 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Nade VS, Dwivedi S, Kawale LA, et al. Effect of Hibiscus rosa sinensis on reserpine-induced neurobehavioral and biochemical alterations in rats. Indian J Exp Biol 2009; 47: 559–563. [PubMed] [Google Scholar]
  • 107.Begum Z, Younus I. Hibiscus rosa sinensis mediate anxiolytic effect via modulation of ionotropic GABA-A receptors: possible mechanism of action. Metab Brain Dis 2018; 33: 823–827. [DOI] [PubMed] [Google Scholar]
  • 108.Pichiah PB, Cho SH, Han SK, et al. Fermented barley supplementation modulates the expression of hypothalamic genes and reduces energy intake and weight gain in rats. J Med Food 2016; 19: 418–426. [DOI] [PubMed] [Google Scholar]
  • 109.Wu J, Tang C, Yao S, et al. Anti-inflammatory inositol derivatives from the whole plant of Inula cappa. J Nat Prod 2015; 78: 2332–2338. [DOI] [PubMed] [Google Scholar]
  • 110.Kalola J, Shah R, Patel A, et al. Anti-inflammatory and immunomodulatory activities of Inula cappa roots (Compositae). J Complement Integr Med 2017. DOI: /j/jcim.2017.14.issue-3/jcim-2016-0083/jcim-2016-0083.xml. [DOI] [PubMed] [Google Scholar]
  • 111.Cho YC, Kim BR, Le HTT, et al. Antiinflammatory effects on murine macrophages of ethanol extracts of Lygodium japonicum spores via inhibition of NFkappaB and p38. Mol Med Rep 2017; 16: 4362–4370. [DOI] [PubMed] [Google Scholar]
  • 112.Lin KH, Yang YY, Yang CM, et al. Antioxidant activity of herbaceous plant extracts protect against hydrogen peroxide-induced DNA damage in human lymphocytes. BMC Res Notes 2013; 6: 490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tian W, Akanda MR, Islam A, et al. The anti-stress effect of Mentha arvensis in immobilized rats. Int J Mol Sci 2018; 19: 355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Verma SM, Arora H, Dubey R. Anti-inflammatory and sedative – hypnotic activity of the methanolic extract of the leaves of Mentha arvensis. Anc Sci Life 2003; 23: 95–99. [PMC free article] [PubMed] [Google Scholar]
  • 115.Oinonen PP, Jokela JK, Hatakka AI, et al. Linarin, a selective acetylcholinesterase inhibitor from Mentha arvensis. Fitoterapia 2006; 77: 429–434. [DOI] [PubMed] [Google Scholar]
  • 116.Feng X, Wang X, Liu Y, et al. Linarin inhibits the acetylcholinesterase activity in-vitro and ex-vivo. Iran J Pharm Res 2015; 14: 949–954. [PMC free article] [PubMed] [Google Scholar]
  • 117.Rawat S, Jugran A, Giri L, et al. Assessment of antioxidant properties in fruits of Myrica esculenta: A popular wild edible species in Indian Himalayan region. Evid-Based Complem Alt Med 2011; 2011: 512787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Agnihotri S, Wakode S, Ali M. Essential oil of Myrica esculenta Buch. Ham.: Composition, antimicrobial and topical anti-inflammatory activities. Nat Prod Res 2012; 26: 2266–2269. [DOI] [PubMed] [Google Scholar]
  • 119.Michael JS, Kalirajan A, Padmalatha C, et al. In vitro antioxidant evaluation and total phenolics of methanolic leaf extracts of Nyctanthes arbor-tristis L. Chinese J Nat Med 2013; 11: 484–487. [DOI] [PubMed] [Google Scholar]
  • 120.Agrawal J, Pal A. Nyctanthes arbor-tristis Linn – a critical ethnopharmacological review. J Ethnopharmacol 2013; 146: 645–658. [DOI] [PubMed] [Google Scholar]
  • 121.Puri A, Saxena R, Saxena RP, et al. Immunostimulant activity of Nyctanthes arbor-tristis L. J Ethnopharmacol 1994; 42: 31–37. [DOI] [PubMed] [Google Scholar]
  • 122.Bharshiv CK, Garg SK, Bhatia AK. Immunomodulatory activity of aqueous extract of Nyctanthes arbor-tristis flowers with particular reference to splenocytes proliferation and cytokines induction. Indian J Pharmacol 2016; 48: 412–417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kakoti BB, Pradhan P, Borah S, et al. Analgesic and anti-inflammatory activities of the methanolic stem bark extract of Nyctanthes arbor-tristis linn. BioMed Res Int 2013; 2013: 826295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Saxena RS, Gupta B, Lata S. Tranquilizing, antihistaminic and purgative activity of Nyctanthes arbor tristis leaf extract. J Ethnopharmacol 2002; 81: 321–325. [DOI] [PubMed] [Google Scholar]
  • 125.Balanehru S, Nagarajan B. Intervention of Adriamycin induced free radical damage. Biochem Int 1992; 28: 735–744. [PubMed] [Google Scholar]
  • 126.Archana R, Namasivayam A. Effect of Ocimum sanctum on noise induced changes in neutrophil functions. J Ethnopharmacol 2000; 73: 81–85. [DOI] [PubMed] [Google Scholar]
  • 127.Sakina MR, Dandiya PC, Hamdard ME, et al. Preliminary psychopharmacological evaluation of Ocimum sanctum leaf extract. J Ethnopharmacol 1990; 28: 143–150. [DOI] [PubMed] [Google Scholar]
  • 128.Sembulingam K, Sembulingam P, Namasivayam A. Effect of Ocimum sanctum Linn on the changes in central cholinergic system induced by acute noise stress. J Ethnopharmacol 2005; 96: 477–482. [DOI] [PubMed] [Google Scholar]
  • 129.Jothie Richard E, Illuri R, Bethapudi B, et al. Anti-stress activity of Ocimum sanctum: Possible effects on hypothalamic-pituitary-adrenal axis. Phytother Res 2016; 30: 805–814. [DOI] [PubMed] [Google Scholar]
  • 130.Pal M, Bhardwaj A, Manickam M, et al. Protective efficacy of the caterpillar mushroom, Ophiocordyceps sinensis (Ascomycetes), from India in neuronal hippocampal cells against hypoxia. Int J Med Mushrooms 2015; 17: 829–840. [DOI] [PubMed] [Google Scholar]
  • 131.Zhu JS, Halpern GM, Jones K. The scientific rediscovery of an ancient Chinese herbal medicine: Cordyceps sinensis part I. J Altern Complement Med 1998; 4: 289–303. [DOI] [PubMed] [Google Scholar]
  • 132.Zhu JS, Halpern GM, Jones K. The scientific rediscovery of a precious ancient Chinese herbal regimen: Cordyceps sinensis: Part II. J Altern Complement Med 1998; 4: 429–457. [DOI] [PubMed] [Google Scholar]
  • 133.D’Souza JJ, D’Souza P P, Fazal F, et al. Anti-diabetic effects of the Indian indigenous fruit Emblica officinalis Gaertn: Active constituents and modes of action. Food Funct 2014; 5: 635–644. [DOI] [PubMed] [Google Scholar]
  • 134.Yang B, Liu P. Composition and biological activities of hydrolyzable tannins of fruits of Phyllanthus emblica. J Agric Food Chem 2014; 62: 529–541. [DOI] [PubMed] [Google Scholar]
  • 135.Variya BC, Bakrania AK, Patel SS. Emblica officinalis (Amla): A review for its phytochemistry, ethnomedicinal uses and medicinal potentials with respect to molecular mechanisms. Pharmacol Res 2016; 111: 180–200. [DOI] [PubMed] [Google Scholar]
  • 136.Golechha M, Sarangal V, Ojha S, et al. Anti-inflammatory effect of Emblica officinalis in rodent models of acute and chronic inflammation: Involvement of possible mechanisms. Int J Inflam 2014; 2014: 178408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Golechha M, Bhatia J, Ojha S, et al. Hydroalcoholic extract of Emblica officinalis protects against kainic acid-induced status epilepticus in rats: Evidence for an antioxidant, anti-inflammatory, and neuroprotective intervention. Pharm Biol 2011; 49: 1128–1136. [DOI] [PubMed] [Google Scholar]
  • 138.Vasudevan M, Parle M. Memory enhancing activity of Anwala churna (Emblica officinalis Gaertn.): An Ayurvedic preparation. Physiol Behav 2007; 91: 46–54. [DOI] [PubMed] [Google Scholar]
  • 139.Zhang K, Ge Z, Da Y, et al. Plumbagin suppresses dendritic cell functions and alleviates experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 273: 42–52. [DOI] [PubMed] [Google Scholar]
  • 140.Jia Y, Jing J, Bai Y, et al. Amelioration of experimental autoimmune encephalomyelitis by plumbagin through down-regulation of JAK-STAT and NF-kappaB signaling pathways. PloS ONE 2011; 6: e27006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Bopaiah CP, Pradhan N. Central nervous system stimulatory action from the root extract of Plumbago zeylanica in rats. Phytother Res 2001; 15: 153–156. [DOI] [PubMed] [Google Scholar]
  • 142.George BP, Parimelazhagan T, Kumar YT, et al. Antitumor and wound healing properties of Rubus ellipticus Smith. J Acupunct Merid Studies 2015; 8: 134–141. [DOI] [PubMed] [Google Scholar]
  • 143.Sharma US, Kumar A. In vitro antioxidant activity of Rubus ellipticus fruits. J Adv Pharm Technol Res 2011; 2: 47–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kumar V, Van Staden J. A review of Swertia chirayita (Gentianaceae) as a traditional medicinal plant. Front Pharmacol 2015; 6: 308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Li J, Zhao YL, Huang HY, et al. Phytochemistry and pharmacological activities of the genus Swertia (Gentianaceae): A review. Am J Chinese Med 2017; 45: 667–736. [DOI] [PubMed] [Google Scholar]
  • 146.Banerjee S, Sur TK, Mandal S, et al. Assessment of the anti-inflammatory effects of Swertia chirata in acute and chronic experimental models in male albino rats Indian J Pharmacol 2000; 32: 21–24. [Google Scholar]
  • 147.Das CS, Bhadra S, Roy S, et al. Analgesic and anti-inflammatory activities of ethanolic root extract of Swertia chirata (Gentianaceae). Jordan J Biol Sci 2012; 5: 31–36. [Google Scholar]
  • 148.Sukprasansap M, Chanvorachote P, Tencomnao T. Cleistocalyx nervosum var. paniala berry fruit protects neurotoxicity against endoplasmic reticulum stress-induced apoptosis. Food Chem Toxicol 2017; 103: 279–288. [DOI] [PubMed] [Google Scholar]
  • 149.Khan A, Nazar H, Sabir SM, et al. Antioxidant activity and inhibitory effect of some commonly used medicinal plants against lipid per-oxidation in mice brain. African J Trad Complem Alt Med 2014; 11: 83–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Park JH, Joo HS, Yoo KY, et al. Extract from Terminalia chebula seeds protect against experimental ischemic neuronal damage via maintaining SODs and BDNF levels. Neurochem Res 2011; 36: 2043–2050. [DOI] [PubMed] [Google Scholar]
  • 151.Huebner FR, Lieberman KW, Rubino RP, et al. Demonstration of high opioid-like activity in isolated peptides from wheat gluten hydrolysates. Peptides 1984; 5: 1139–1147. [DOI] [PubMed] [Google Scholar]
  • 152.Pascual J, Leno C. A woman with daily headaches. J Headache Pain 2005; 6: 91–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Umamaheswari M, Asokkumar K, Umamageswari N, et al. Protective effect of the leaves of Vitex negundo against ethanol-induced cerebral oxidative stress in rats. Tanzania J Health Res 2012; 14: 21–28. [DOI] [PubMed] [Google Scholar]
  • 154.Zheng CJ, Li HQ, Ren SC, et al. Phytochemical and pharmacological profile of Vitex negundo. Phytother Res 2015; 29: 633–647. [DOI] [PubMed] [Google Scholar]
  • 155.Chan EWC, Wong SK, Chan HT. Casticin from Vitex species: A short review on its anticancer and anti-inflammatory properties. J Integrat Med 2018; 16: 147–152; [DOI] [PubMed] [Google Scholar]
  • 156.Wazir A, Mehjabeen, Jahan N, et al. Antibacterial, antifungal and antioxidant activities of some medicinal plants. Pak J Pharm Sci 2014; 27: 2145–2152. [PubMed] [Google Scholar]
  • 157.Ahmed S, Shakeel F. . Voltammetric determination of antioxidant character in Berberis lycium Royel, Zanthoxylum armatum and Morus nigra Linn plants. Pak J Pharm Sci 2012; 25: 501–507. [PubMed] [Google Scholar]
  • 158.Nooreen Z, Kumar A, Bawankule DU, et al. New chemical constituents from the fruits of Zanthoxylum armatum and its in vitro anti-inflammatory profile. Nat Prod Res 2017: 1–8. [DOI] [PubMed] [Google Scholar]
  • 159.Guo T, Deng YX, Xie H, et al. Antinociceptive and anti-inflammatory activities of ethyl acetate fraction from Zanthoxylum armatum in mice. Fitoterapia 2011; 82: 347–351. [DOI] [PubMed] [Google Scholar]
  • 160.Choi DJ, Kim SL, Choi JW, et al. Neuroprotective effects of corn silk maysin via inhibition of H2O2-induced apoptotic cell death in SK-N-MC cells. Life Sci 2014; 109: 57–64. [DOI] [PubMed] [Google Scholar]
  • 161.Maghbooli M, Golipour F, Moghimi Esfandabadi A, et al. Comparison between the efficacy of ginger and sumatriptan in the ablative treatment of the common migraine. Phytother Res 2014; 28: 412–415. [DOI] [PubMed] [Google Scholar]
  • 162.Semwal RB, Semwal DK, Combrinck S, et al. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015; 117: 554–568. [DOI] [PubMed] [Google Scholar]
  • 163.Otunola GA, Oloyede OB, Oladiji AT, et al. Selected spices and their combination modulate hypercholesterolemia-induced oxidative stress in experimental rats. Biol Res 2014; 47: 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Ho SC, Chang KS, Lin CC. Anti-neuroinflammatory capacity of fresh ginger is attributed mainly to 10-gingerol. Food Chem 2013; 141: 3183–3191. [DOI] [PubMed] [Google Scholar]
  • 165.Deng XY, Xue JS, Li HY, et al. Geraniol produces antidepressant-like effects in a chronic unpredictable mild stress mice model. Physiol Behav 2015; 152: 264–271. [DOI] [PubMed] [Google Scholar]
  • 166.Grzanna R, Phan P, Polotsky A, et al. Ginger extract inhibits beta-amyloid peptide-induced cytokine and chemokine expression in cultured THP-1 monocytes. J Alt Complem Med 2004; 10: 1009–1013. [DOI] [PubMed] [Google Scholar]
  • 167.Jafarzadeh A, Arabi Z, Ahangar-Parvin R, et al. Ginger extract modulates the expression of chemokines CCL20 and CCL22 and their receptors (CCR6 and CCR4) in the central nervous system of mice with experimental autoimmune encephalomyelitis. Drug Res 2017; 67: 632–639. [DOI] [PubMed] [Google Scholar]
  • 168.Park G, Kim HG, Ju MS, et al. 6-Shogaol, an active compound of ginger, protects dopaminergic neurons in Parkinson’s disease models via anti-neuroinflammation. Acta Pharmacol Sinica 2013; 34: 1131–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Lim S, Moon M, Oh H, et al. Ginger improves cognitive function via NGF-induced ERK/CREB activation in the hippocampus of the mouse. J Nutr Biochem 2014; 25: 1058–1065. [DOI] [PubMed] [Google Scholar]
  • 170.Qian QH, Yue W, Chen WH, et al. Effect of gingerol on substance P and NK1 receptor expression in a vomiting model of mink. Chinese Med J 2010; 123: 478–484. [PubMed] [Google Scholar]
  • 171.Gauthier ML, Beaudry F, Vachon P. Intrathecal [6]-gingerol administration alleviates peripherally induced neuropathic pain in male Sprague-Dawley rats. Phytother Res 2013; 27: 1251–1254. [DOI] [PubMed] [Google Scholar]
  • 172.Oboh G, Ademiluyi AO, Akinyemi AJ. Inhibition of acetylcholinesterase activities and some pro-oxidant induced lipid peroxidation in rat brain by two varieties of ginger (Zingiber officinale). Experim Toxicol Pathol 2012; 64: 315–319. [DOI] [PubMed] [Google Scholar]
  • 173.Xue J, Li H, Deng X, et al. L-Menthone confers antidepressant-like effects in an unpredictable chronic mild stress mouse model via NLRP3 inflammasome-mediated inflammatory cytokines and central neurotransmitters. Pharmacol Biochem Behav 2015; 134: 42–48. [DOI] [PubMed] [Google Scholar]
  • 174.Chuchawankul S, Khorana N, Poovorawan Y. Piperine inhibits cytokine production by human peripheral blood mononuclear cells. Genet Mol Res 2012; 11: 617–627. [DOI] [PubMed] [Google Scholar]
  • 175.Ahmad MP, Hussain A, Siddiqui HH, et al. Effect of methanolic extract of Asparagus racemosus Willd. on lipopolysaccharide induced-oxidative stress in rats. Pakistan J Pharmaceut Sci 2015; 28: 509–513. [PubMed] [Google Scholar]
  • 176.Singh GK, Garabadu D, Muruganandam AV, et al. Antidepressant activity of Asparagus racemosus in rodent models. Pharmacol Biochem Behav 2009; 91: 283–290. [DOI] [PubMed] [Google Scholar]
  • 177.Krishnamurthy S, Garabadu D, Reddy NR. Asparagus racemosus modulates the hypothalamic-pituitary-adrenal axis and brain monoaminergic systems in rats. Nutr Neurosci 2013; 16: 255–261. [DOI] [PubMed] [Google Scholar]
  • 178.Meena J, Ojha R, Muruganandam AV, et al. Asparagus racemosus competitively inhibits in vitro the acetylcholine and monoamine metabolizing enzymes. Neurosci Lett 2011; 503: 6–9. [DOI] [PubMed] [Google Scholar]
  • 179.Ojha R, Sahu AN, Muruganandam AV, et al. Asparagus racemosus enhances memory and protects against amnesia in rodent models. Brain Cognit 2010; 74: 1–9. [DOI] [PubMed] [Google Scholar]
  • 180.Fatima N, Rahman MM, Khan MA, et al. A review on Ipomoea carnea: Pharmacology, toxicology and phytochemistry. J Complem Integrat Med 2014; 11: 55–62. [DOI] [PubMed] [Google Scholar]
  • 181.Press JR, Shrestha KK, Sutton DA. Annotated checklist of the flowering plants of Nepal http://www.efloras.org/flora_page.aspx?flora_id=110 (accessed 3 April 2018).
  • 182.Baba Ramdev Health Products. Banphool Oil, http://www.ramdevproducts.com/banphool-oil.htm (2018, accessed 1 April 2018).
  • 183.Kaur M, Prakash A, Kalia AN. Neuroprotective potential of antioxidant potent fractions from Convolvulus pluricaulis Chois. in 3-nitropropionic acid challenged rats. Nutr Neurosci 2016; 19: 70–78. [DOI] [PubMed] [Google Scholar]
  • 184.Agarwa P, Sharma B, Fatima A, et al. An update on Ayurvedic herb Convolvulus pluricaulis Choisy. Asian Pac J Trop Biomed 2014; 4: 245–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Sharma K, Bhatnagar M, Kulkarni SK. Effect of Convolvulus pluricaulis Choisy and Asparagus racemosus Willd on learning and memory in young and old mice: A comparative evaluation. Indian J Exp Biol 2010; 48: 479–485. [PubMed] [Google Scholar]
  • 186.Heba M, Faraz S, Banerjee S. Effect of Shankhpushpi on alcohol addiction in mice. Pharmacogn Mag 2017; 13: S148–S153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Bihaqi SW, Singh AP, Tiwari M. Supplementation of Convolvulus pluricaulis attenuates scopolamine-induced increased tau and amyloid precursor protein (AbetaPP) expression in rat brain. Indian J Pharmacol 2012; 44: 593–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Malik J, Choudhary S, Kumar P. Protective effect of Convolvulus pluricaulis standardized extract and its fractions against 3-nitropropionic acid-induced neurotoxicity in rats. Pharm Biol 2015; 53: 1448–1457. [DOI] [PubMed] [Google Scholar]
  • 189.Malik J, Karan M, Vasisht K. Nootropic, anxiolytic and CNS-depressant studies on different plant sources of shankhpushpi. Pharm Biol 2011; 49: 1234–1242. [DOI] [PubMed] [Google Scholar]
  • 190.Bihaqi SW, Sharma M, Singh AP, et al. Neuroprotective role of Convolvulus pluricaulis on aluminium induced neurotoxicity in rat brain. J Ethnopharmacol 2009; 124: 409–415. [DOI] [PubMed] [Google Scholar]
  • 191.Flowers of India. Kewda, http://www.flowersofindia.net/catalog/slides/Kewda.html (2018, accessed 28 May 2018).
  • 192.Rajeswari J, Kesavan K, Jayakar B. Phytochemical and pharmacological evaluation of prop roots of Pandanus fascicularis Lam. Asian Pac J Trop Med 2011; 4: 649–653. [DOI] [PubMed] [Google Scholar]
  • 193.Okugawa H, Ueda R, Matsumoto K, et al. Effect of alpha-santalol and beta-santalol from sandalwood on the central nervous system in mice. Phytomedicine 1995; 2: 119–126. [DOI] [PubMed] [Google Scholar]
  • 194.d' Avila Farias M, Oliveira PS, Dutra FS, et al. Eugenol derivatives as potential anti-oxidants: Is phenolic hydroxyl necessary to obtain an effect? J Pharm Pharmacol 2014; 66: 733–746. [DOI] [PubMed] [Google Scholar]
  • 195.Halder S, Mehta AK, Kar R, et al. Clove oil reverses learning and memory deficits in scopolamine-treated mice. Planta Med 2011; 77: 830–834. [DOI] [PubMed] [Google Scholar]
  • 196.Ardjmand A, Fathollahi Y, Sayyah M, et al. Eugenol depresses synaptic transmission but does not prevent the induction of long-term potentiation in the CA1 region of rat hippocampal slices. Phytomedicine 2006; 13: 146–151. [DOI] [PubMed] [Google Scholar]
  • 197.Liu BB, Luo L, Liu XL, et al. Essential oil of Syzygium aromaticum reverses the deficits of stress-induced behaviors and hippocampal p-ERK/p-CREB/brain-derived neurotrophic factor expression. Planta Med 2015; 81: 185–192. [DOI] [PubMed] [Google Scholar]
  • 198.Jeon SJ, Kim E, Lee JS, et al. Maslinic acid ameliorates NMDA receptor blockade-induced schizophrenia-like behaviors in mice. Neuropharmacology 2017; 126: 168–178. [DOI] [PubMed] [Google Scholar]
  • 199.Hartnoll G, Moore D, Douek D. Near fatal ingestion of oil of cloves. Arch Dis Child 1993; 69: 392–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Lane BW, Ellenhorn MJ, Hulbert TV, et al. Clove oil ingestion in an infant. Hum Exp Toxicol 1991; 10: 291–294. [DOI] [PubMed] [Google Scholar]
  • 201.Soubir T. Antioxidant activities of some local bangladeshi fruits (Artocarpus heterophyllus, Annona squamosa, Terminalia bellirica, Syzygium samarangense, Averrhoa carambola and Olea europa). Sheng Wu Gong Cheng Xue Bao 2007; 23: 257–261. [PubMed] [Google Scholar]
  • 202.Chalise JP, Acharya K, Gurung N, et al. Antioxidant activity and polyphenol content in edible wild fruits from Nepal. Int J Food Sci Nutr 2010; 61: 425–432. [DOI] [PubMed] [Google Scholar]
  • 203.Tanaka M, Kishimoto Y, Saita E, et al. Terminalia bellirica extract inhibits low-density lipoprotein oxidation and macrophage inflammatory response in vitro. Antioxidants 2016; 5: 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Jayesh K, Helen LR, Vysakh A, et al. Ethyl acetate fraction of Terminalia bellirica (Gaertn.) Roxb. fruits inhibits proinflammatory mediators via down regulating nuclear factor-kappaB in LPS stimulated Raw 264.7 cells. Biomed Pharmacother 2017; 95: 1654–1660. [DOI] [PubMed] [Google Scholar]
  • 205.Dhingra D, Valecha R. Evaluation of antidepressant-like activity of aqueous and ethanolic extracts of Terminalia bellirica Roxb. fruits in mice. Indian J Exp Biol 2007; 45: 610–616. [PubMed] [Google Scholar]
  • 206.Manandhar K, Risal A, Linde M, et al. The burden of headache disorders in Nepal: Estimates from a population-based survey. J Headache Pain 2015; 17: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Kunwar RM, Mahat L, Acharya RP, et al. Medicinal plants, traditional medicine, markets and management in far-west Nepal. J Ethnobiol Ethnomed 2013; 9: 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Kristoffersen ES, Lundqvist C. Medication-overuse headache: A review. J Pain Res 2014; 7: 367–378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Diener HC, Dodick D, Evers S, et al. Pathophysiology, prevention, and treatment of medication overuse headache. Lancet Neurol 2019; 18: 891–902. [DOI] [PubMed] [Google Scholar]
  • 210.Nepalese Ministry of Health and Population. Nepal population report 2011. Kathmandu: Government of Nepal, 2011, pp.1–186. [Google Scholar]
  • 211.Risal A, Manandhar K, Steiner TJ, et al. Estimating prevalence and burden of major disorders of the brain in Nepal: Cultural, geographic, logistic and philosophical issues of methodology. J Headache Pain 2014; 15: 51. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cephalalgia are provided here courtesy of SAGE Publications

RESOURCES