Open access peer-reviewed chapter - ONLINE FIRST

Effects of Acupuncture Treatment on Polycystic Ovary Syndrome (PCOS)

Written By

Gunasekaran Ramanathan

Submitted: 24 August 2023 Reviewed: 20 October 2023 Published: 19 December 2023

DOI: 10.5772/intechopen.113799

Polycystic Ovary Syndrome - Symptoms, Causes and Treatment IntechOpen
Polycystic Ovary Syndrome - Symptoms, Causes and Treatment Edited by Zhengchao Wang

From the Edited Volume

Polycystic Ovary Syndrome - Symptoms, Causes and Treatment [Working Title]

Dr. Zhengchao Wang

Chapter metrics overview

37 Chapter Downloads

View Full Metrics

Abstract

Polycystic ovarian syndrome (PCOS) is an imbalance of endocrine hormones with a group of symptoms that occur in the reproductive age of females. It is diagnosed by androgen excess and ovarian dysfunction. Many pharmacological-based drugs and treatments are available, however, the effectiveness of existing therapies is less. Yet no cure for PCOS, but symptoms can be managed with medications and lifestyle modifications. Hence, an alternative treatment method is needed for a complete cure. Currently, much clinical evidence has revealed that Acupuncture (AP) and Electro-acupuncture treatment (EAT) are effective treatments for regulating reproductive hormone levels, normalizing testosterone, reducing ovarian cysts, enhancing ovulation, and weakening insulin resistance in normal and obese women with PCOS. Further, β-endorphin has a more crucial role in PCO, AP, or EAT, which may alter the formation of β-endorphin. The mechanism of AP and EAT for PCOS has not been widely reviewed so far. A good understanding of the AP and EAT would be helpful in women with PCOS. This chapter aimed to overview the probable mechanisms and experimental evidence-based data of acupuncture treatment on PCOS in animal models and human patients. We hope that the chapter study will contribute to a better understanding of the PCOS and AP treatment.

Keywords

  • acupuncture
  • electro-acupuncture treatment
  • PCOS
  • anovulation
  • ovarian cysts anovulation
  • HR variability

1. Introduction

Polycystic ovary syndrome (PCOS) is an endocrine disorder. In the modern lifestyle, the disorder affects many reproductive-age women worldwide. In 2010 data worldwide, 116 million (3.42%) women were affected by PCOS [1]. PCO is majorly associated with the dysfunction of ovaries. Based on studies, many factors are involved. The clinical signs of PCO are hyperandrogenism, ovulatory dysfunction, metabolic dysfunction, and obesity [2, 3] further, it is associated with cardiovascular disease CVD, cancer, diabetes, and endometrial dysfunction [4, 5], but the exact cause, etiology, and pathophysiology are not clearly understood. Yet there is no perfect treatment for all diagnosed with PCOS [6, 7]. Depending on the patient, the PCO management method and selection of the best therapy option and their priorities vary [8, 9].

The current management, therapy, and pharmacological medications are less effective in PCOS, yet some complicated women with PCOS are left untreated [10]. Currently, the pharmacological therapy for women with PCOS is using an oral estrogen receptor modulator, but it’s 40–50% ineffective in many cases. It’s linked with side effects such as breast tenderness, headaches, mood swings, arthralgias, myalgias, and fatigue [11, 12]. Further studies showed pituitary Gonadotropin (GnH) hormone is effective in inducing ovulation; however, it causes overstimulation syndrome and the development of multiple follicles [13].

Recent research showed that complementary and alternative medicine (CAM) as alternative therapy might benefit the management of PCOS [14]. The CAM therapeutic input has classes of physical, nutritional, and psychological, or all in combination [15]. Hence, clear-cut PCOS pathogenesis and perfect management are urgently needed. In this book chapter, we update the pathogenesis and current management, particularly the beneficial effects of electro-acupuncture on PCOS.

Advertisement

2. What is acupuncture treatment?

Acupuncture has origins in traditional Chinese medicine (TCM) [16], More than 3000 years, ago acupuncture treatment was used; it’s a basic part of CAM [14] acupuncture is a specific type of sensory stimulation in the skin and muscles using thin needles. It may be useful to treat some chronic pain, and some abnormal physical conditions, and as a complementary treatment for many disorders [17].

Advertisement

3. How is acupuncture working?

TCM describes that a person’s health results from a tuneful balance of the complementary extremes of Chinese Yin and Yang of the life force known as Qi marked “chi.” It believes that disease is the result of an imbalance of these forces. In the human body, the Qi flows through specific pathways, or meridians. These energy flows and pathways are accessible through specific acupuncture points. In the human body 361, acupuncture points have been identified. Inserting selected thin needles into these suitable acupoints with suitable combinations will bring the flow of energy back into balance [18].

Stimulation of many acupuncture points affects multiple sensory neuron activity. These points are called the receptive fields. The physical insertion of a thin needle may affect pain processing in the muscles and central nervous system (CNS), further increasing blood flow to certain organs and parts of the body [19]. However, the exact mechanism of action of acupuncture is not clear.

Advertisement

4. What’s electro-acupuncture treatment (EAT)?

In China1950s EAT was developed for its precise use in surgical operations. In the early days, acupuncture was used for analgesia; while using it, the anesthetists had to rotate the needles manually throughout the surgery, which was difficult, so they developed an electro-acupuncture (EA) apparatus. EAT, in which acupuncture is united with electrical stimulation. Initially, EA analgesia was used largely in China, and interest showed in the West. It’s useful to reduce the use of analgesic drugs during surgery. However, EA in practice is personal and depends on their experience. Some practitioners said EA is against the spirit of ‘natural’ acupuncture therapy, but others said EAT is good for patients with chronic nociceptive pain and good for drug addiction.

The analgesic effects of EAT are more effective than those of the manual acupuncture method. An fMRI study of normal adults found that stimulation with EA at a frequency of 3 Hz on acupuncture point LI-4 Hegu produced fMRI signal increases in the precentral gyrus, postcentral gyrus/inferior parietal lobule, and putamen/insula [20], whereas normal acupuncture through specific needle manipulation manually of the same acupuncture point produced decreases in fMRI signals in the putamen/insula, posterior cingulate, and superior temporal gyrus [21]. EA also increases the tissue content of endorphins [22].

Electro-acupuncture (EA) can be used for seizures, dizziness, aphasia, headache, chorea, stroke, drug addiction, PCO, and many more. Currently, two modes of EA are used commonly: high-intensity high-frequency (50 to 200 Hz) and low-intensity low-frequency (1 to 4 Hz). In this EA therapy, instead of hand stimulation through a needle, electro-stimulation is used to treat the diseases. In EAT-specific needles are placed in the scalp (anastomotic linkage of lymphatic drainage) and connected with an electro-acupuncture machine to stimulate the acupuncture points. The pulse duration, intensity, and frequency of the electric current must be altered depending on the patient. EAT stabilizes the treatment through its fast rotation; we can stimulate more than two points simultaneously and control the current level depending on the patient. Usually, EAT duration is 10 to 20 minutes per time for 7–10 days depending on the disease.

EAT should be used with caution. Use the needle in the right position, not bend or break. When the intensity of the current used for acupuncture points close to the brain area, medulla, lateral line of the occipital area, or other areas, at all times the right amount of current must be used. Do not increase the current suddenly to a large amount because it may cause an increase in heart rate (HR) and breathing stops. It’s contraindicated for cancer, heart diseases, and brain diseases.

Advertisement

5. Electroacupuncture treatment for PCOS

Conventional therapy (first-line) is not yet satisfactory, but the nonpharmacological therapy of normal acupuncture and EA has become popular. Based on multiple clinical trials, both manual acupuncture and EAT have a good effect on treating women with PCO [23, 24, 25, 26, 27]. Earlier studies and clinical trials suggested that proper acupuncture treatment improves menstruation and ovulation [2829]. It has also been reported that EAT decreases testosterone levels and improves insulin sensitivity in animals and humans with PCOS [23]. Further, it regulates the levels of the anti-Müllerian hormone (AMH) and hypothalamic–pituitary-ovarian (HPO) axis. A recent animal models PCO study explain that acupuncture could improve insulin resistance inhibit the PI3K/AKT/mTOR pathway, or activate the protein kinase (AMPK) pathway [30]. Further, it reduces anxiety and improves health-related quality of life [31]. Acupuncture treatment for patients with PCOS decreases the abnormally increased luteinizing hormone (LH) [29, 32] and increased testosterone [33] without side effects; it’s less expensive treatment [34]. Collectively, the manual or EAT for PCOs improves the pregnancy rate. Ovulation rate, sexual hormonal imbalance, metabolic dysfunction, and insulin resistance (Figure 1).

Figure 1.

Manual or EAT improves PCOS-related symptoms such as pregnancy rate, ovulation rate, sexual hormonal imbalance, metabolic dysfunction, and insulin resistance through different biological mechanisms.

Advertisement

6. Methods of electroacupuncture treatment

Stimulation through EA is more effective and can be easily quantifiable compared to manual acupuncture, further reducing the bias. Earlier studies showed Zigong (EX-CA 1), Sanyinjiao (SP 6), Zhongji (CV 3), and Guanyuan (BL 26) acupuncture points were chosen for the formula. In addition to that, three arbitrary acupuncture points have been selected based on phases of the menstrual cycle: 1) after menstruation, using acupuncture points for the needle: Taixi (KI 3) and Taichong (LR 3). 2) In the ovulatory period, using acupuncture points: Xuehai (SP 10) and Mingmen (CV 4). 3) Using acupuncture points before the beginning of the next menstruation: Geshu (BL 17) and Xuehai (SP 10).

The broadly used acupuncture point was SP6 (Sanyinjiao) for reproductive problems, nourishing organs, and activating blood; the second ST29 (Guilai) acupoint was used for reducing pain and regulating circulation and menstruation. An EAT, or manual acupuncture needle stimulation, induces and enhances muscle conduction and various chemical signals to stimulate the central release of various factors through sympathetic nerve conduction, which is believed to regulate the woman’s reproductive axis (Figure 2).

Figure 2.

The most of the acupuncture needle points are on the abdominal area for human and animal for PCO, the acupuncture point SP6 (Sanyinjiao)for reproductive problem, nourishing organs, and activating blood. The acupuncture point ST29 (Guilai) was used for reducing pain and regulate menstruation.

Advertisement

7. Needle using

An earlier study [35] showed that each acupuncture point needled used a filiform needle (length 25–50 mm and diameter 0.25–026 mm) to attain a Deqi sensation3 (at the needling acupuncture point, feel a sensation like numbness and swelling, and it radiates along the reliable meridian). Needled used in each acupuncture point will be a filiform needle (length 25–50 mm and diameter 0.25 mm) to achieve a Deqi sensation. Then, an auxiliary needle (length: 13 mm and diameter: 0.18 mm) will be inserted to a depth of 2 mm and 2 mm lateral to the first needle without achieving Deqi.4. With the help of an EA instrument, fixed or required electrical current will be given at the acupuncture point, with one electrode to the auxiliary and the other electrode to the filiform needle [35].

Duration of treatment: Electrical stimulation for a minimum of 30 min for each session. From the 50th day of menstruation three times per week or withdrawal bleeding until before the next menstruation for three menstrual cycles.

Expected outcome: Primary outcome: the ovulation rate. Secondary outcomes: ovulation time point, endometrial thickness, dominant follicle rate, and follicular size Hormone level: FSH, LH, GnRH, Estradiol (E2) level, and pregnancy rate.

Advertisement

8. EAT rectifies the anovulation in PCO

8.1 Experimental evidence

Experimental evidence PCOS is a complex metabolic and endocrine disorder [36]. In the reproductive age of females, many cases of anovulation occur, which leads to infertility. EA treatment is a safe and effective treatment for anovulation in women 5A suitable EA treatment may effectively reduce the number of ovarian cysts, reduce ovarian volume, and enhance blood flow to the ovaries. Further, it reverses the insulin levels and insulin sensitivity back to normal, which reduces the blood glucose level. Further, it controls the sympathetic activity to normal [37]. Only very few animal model studies are available on the effect of EAT on the sympathetic activity in PCOS. Hence, in our laboratory, we aimed to assess the effect of EAT on anovulation and increased sympathetic nerve activity in EV-induced PCO rate models.

EAT: Repeated low-frequency EAT was carried out for 20 minutes for the PCO group for two days up to 8–10 weeks. Just before EAT rats were anesthetized, the needles were inserted into the erector spinae muscle in the somatic segment and bilaterally in the biceps femoris related to the nerve innervations of the ovaries, inserted to a depth of 0.3–0.51 cm. 6 These are connected to the EA machine using the frequencies of 80 Hz (burst frequency) and 2 Hz (frequency low), and until local muscle contraction, the intensity was adjusted to be from 0.8 to 1.31 mA. After the experimental period, animals were anesthetized and the blood samples were collected to measure plasma epinephrine (EPI) and norepinephrine (NE) levels in high-performance liquid chromatography (HPLC).

8.2 Histological study of the ovaries

For the histological study of ovaries, the control and experimental groups were euthanized, ovaries were removed, and the near connective fat tissue was removed. The tissue sections were stained with hematoxylin–eosin (NICE chemicals), and then the slides were used to study the follicles. The slides were viewed under a light microscope, and a well-trained pathologist determined the morphological variation of the normal and abnormal follicles in the follicular population. In this case, a normal ovary has become a polycystic ovary (PCO). We can see on the outer border of the ovary many small fluid-filled sacs present. These sacs are called cysts, which contain immature eggs. The cystic follicle’s failure to release eggs leads to anovulation.

Most of the follicles are cystic in appearance, and stromal hyperplasia was seen. Follicles become atretic; otherwise, the ovary and follicles were considered healthy.

We got the following result: After the experimental period of 30, and 60 days, the normal, EV-induced PCO and EA treatment rates were killed. After 30 days, PCO rates showed a progressive decline in the number of primary and secondary follicles, but after 60 days, there were many morphological variations seen in the PCO rat’s follicles compared to normal healthy follicles, and follicular cysts appeared, big-size atretic follicles, and stromal hyperplasia were also seen. It confirms that the ovary attains PCOS (Figure 3B) similar result was also observed in an earlier study [38]. Repeated EAT after 60 days showed the abnormal follicle morphology back to normal and the number of ovarian cysts considerably reducing (Figure 3C). Furthermore, no considerable morphological differences were seen between the healthy rats ovary (Control group Figure 3A) and the EAT group rats’ ovary (Figure 3).

Figure 3.

Hematoxylin and eosin-stained ovary section. A. The normal control rats ovaries with healthy follicles and corpus luteum, B. EV-induced rat’s ovaries showed cystic appearance, and C. After electroacupuncture treatment in PCOS rats showed reappearance of healthy follicles.

8.3 Epinephrine and norepinephrine levels in rat plasma

In our study, the EV-treated PCO rats showed a significant increase in the epinephrine (EPI) and norepinephrine (NE), but after 60 days of EAT, the PCO group rates showed a significant decrease in both EPI and NE levels (P < 0.001). When it’s compared to the control group with the EAT group, the EPI and NE levels are back to normal (Table 1).

ParametersNormal control ng/ml Mean + SDPCO before EAT ng/ml Mean + SDPCO after EAT ng/ml Mean + SDP value
Epinephrine0.257 + 0.049030.41190 + .039240.26001 + .05191P < 0.001
Norepinephrine0.2386+ 0.057970.3994 + 0.054250.25821 + 0.04945P < 0.001

Table 1.

Epinephrine and norepinephrine level in rat plasma (Normal and PCO, EAT).

Advertisement

9. The potential mechanisms of EAT and anovulation

Chronic anovulation is a main cause of infertility; in both animals and humans, anovulation is the supreme characteristic of PCOS [39]. EAT improves the development and maturation of follicles and oocytes and prevents anovulation [26, 40]. It’s by increasing the expression of P450arom, inhibiting the overexpression of, and upregulating the IRS-1/PI3K/GLUT4 pathway. Further, EA improved in vitro fertilization [41]. Various animal model studies also reported that EA improves ovulation dysfunction by reducing granulosa atrophy, improving follicular arrest, inhibiting the PI3K/AKT/mTOR pathway, and downregulating LncMEG3 expression [42, 43]. EAT also improves the preovulatory follicle number and development of corporalutea. It’s by increasing the hilum blood vessels and blood supply [44].

In women, a range of reproductive hormonal disturbances cause PCO. EAT rectifies the hormonal imbalance of decreased GnRH, estradiol, and increased LH in women with PCO [45]. Acupuncture reduces the abnormal serum sex steroid levels of the adrenal cortex and ovaries. In the PCO animal model study, EAT improved the number of corpora lutea, disturbed estrous cycles, and upregulated the arcuate nucleus kisspeptin protein [46].

Advertisement

10. EAT rectifies the autonomic alteration in women with PCOS

10.1 Experimental evidence

Heart rate (HR) fluctuation is a sign of autonomic imbalance. Based on earlier studies, it’s an initial risk for heart problems, and it may lead to CVD. Further, studies showed that a group of PCOS women has a risk of CVD [4]. Therefore, in our laboratory, we aimed to investigate the autonomic alteration of the HR in women with PCOS and the effect of EAT on it. For this study, we used the estradiol valerate (EV)-induced PCO rat model [47]. In this study, the vaginal smear was taken periodically from the control and experimental group rats. The EV-induced PCO model rat group shows a prolonged diestrus stage compared to the control group; we determine its PCO group rat [47].

Heart rate variability (HRV) analysis: Our laboratory’s heart rate variability (HRV) was assessed in the normal control group and experimental PCO group rats before and after EAT. It’s done from the frequency domain and the time domain analysis. In the time domain analysis, the following data were obtained: mean RR interval, SDNN (standard deviation of RR interval), and rMSSD. For frequency domain analysis, there were two major spectral components: high frequency (HF 0.8–2.5 Hz) and low frequency (LF 0.2–0.8 Hz).

EAT: For this study, we carried out low-frequency EAT in the conformed PCOS rats; each treatment was given once in two days for 15–20 minutes, given up to 4–5 weeks. The EA needles were inserted in the erector spinae muscle in the somatic segment and bilaterally in the biceps femoris related to the nerve innervations of the ovaries, with a needle depth of 0.3–0.5 cm. Then it connected to an EA electrical stimulator with a burst frequency of 80 Hz and a low frequency of 2 Hz.

The result obtained: The EV-induced PCO rate showed a significant reduction (p < 0.05). in the time domain R-R interval, SDNN, and rMSSD compared to the control group, but after 4–5 weeks of EAT, the PCO group rats showed a significant increase in the R-R interval, SDNN, and rMSSD (p < 0.05). It indicates that the repeated low frequency EAT has beneficial effects on PCOS rats; the RR interval and HR significantly returned to normal (Table 2).

ParametersControlPCOEAT
Mean RR176 ± 3.9150 ± 3.9*170.7 ± 3.3**
SDNN13.6 ± 1.510.76 ± 1.5*11.6 ± 0.8
rMSSD5.8 ± 0.93.46 ± 0.69*5.5 ± 0.9**

Table 2.

The control group, PCO, and PCO + EAT groups were compared in time domain indices the values are in mean ± SD.

In comparison with the control group the PCO group showed a significant decrease (p < 0.05)*. And comparison with the PCOS group with PCO + EAT showed a significant increase (p < 0.05)**.

The frequency domain analysis of the PCOS rats showed a significant increase in the low-frequency power LF and LF/ HF ratio. But total power TP (p < 0.05) and high-frequency power HF, when compared to control, significantly decreased. However, after 4–5 weeks of EAT, the LF and LF/HF ratios significantly decreased and the HF and TP increased when compared to untreated PCOS (Table 3).

ParametersControl groupPCO groupPCO+ EAT group
HF15.87 ± 4.048.62 ± 0.7*3.7 ± 2.07**
LF15.07 ± 1.820.32 ± 2.5*16.5 ± 1.7**
LF/HF0.98 ± 0.181.72 ± 0.17*1.2 ± 0.18**
TP71.6 ± 10.341.75 ± 6.9*65.8 ± 11.3**

Table 3.

The frequency domain analysis comparison between groups * and ** indicates significant.

10.2 Mechanisms EAT on HR variability in PCO

In our study, short-term HRV analysis of the EV-induced PCO rat showed a significant decrease in the total power (TP), which is an index of overall HRV. A further significant increase in LF nu showed increased sympathetic activity, and a significantly decreased HF nu indicates decreased parasympathetic activity. Furthermore, the LF/HF ratio also increased, which indicates an imbalance between sympathetic nerve and parasympathetic nerve activity. Earlier studies also showed an increase in LF and LF/HF ratio, which is due to increased sympathetic activity [48]. In another study, EV-induced PCO model rats significantly increased the synthesis of intra-ovarian nerve growth factor (NGF), which is a strong marker for sympathetic activity [49]. Increased activity of sympathetic nerves is also linked with obesity and CVD [50, 51]. In a comparative study, PCOS was treated with exercise with low-frequency EAT, which has shown significantly decreased high muscle sympathetic activity [52]. In the EV-induced PCOS, model rats showed the EAT decreased the high ovarian NGF and high mRNA expression of NGF [37, 53, 54] and adrenergic receptors [55].

10.3 The mechanisms of acupuncture treatment on hyperandrogenism

Hyperandrogenism is a condition in which overproduction of the male sex hormone (testosterone), even though these hormones are present in women at lower levels. But in some women, excess levels of male sex hormones androsterone, androstenedione, and testosterone causes Cushing syndrome, PCO, and malfunction of the adrenal gland. Erlaier’s studies showed that hyperandrogenism is the most important mechanism and clinical feature of PCOS [56]. EAT normalizes the blood testosterone level in women with PCOs [28]. Further, in the rate model PCO, the low-frequency EAT also reduces the serum androgen level through the hypothalamic Oprk1 and Oprm1 central opioid receptors [57]. In another study in PCO women, EAT regulates the AMH and P450arom through this mechanism, it reduces the increased androgen level [58]. A similar result was also observed in the rat model PCO study by Sun et al. [59]. Another PCO animal model study also reveals that acupuncture treatment reduces abnormally increased androgen secretion and reduces the number of androgen receptors [60].

10.4 Obesity and PCO

Obesity has increased the amount of body fat and is a complex disease. It’s the root of many diseases and health problems like diabetes, high cholesterol, heart disease, high blood pressure, and liver disease. Its comorbidity is associated with cardiometabolic dysfunction like hypertension, type 2 diabetes mellitus (T2D), PCOs, and other metabolic syndromes. Increasing women’s body weight and obesity are the main factors in the pathogenesis of PCOS. It is also associated with endometrial carcinoma, hyperinsulinemia, and insulin resistance. PCO is a good example of a metabolic disorder, which also includes insulin resistance and cardiometabolic risk (Figure 4).

Figure 4.

Summary of obesity linked with PCOS.

Earlier studies reported that women being overweight is the reason for PCOs [61, 62, 63]. Conversely, weight loss reduces the risk of reproductive failure, metabolic features of PCOs, and hyperandrogenism [64, 65]. It indicates that treating obesity is the first step to preventing infertility and PCO. Currently, pharmaceutical treatments for obesity are available; however, due to security reasons, there are various limits [66, 67]. Acupuncture treatment is an alternative intervention method for obesity [68]. Some studies suggest that EAT is more effective compared to manual acupuncture, but the effective frequency determination is controversial. In some obesity cases, a combination of diet modification, or exercise, and EAT is essential for weight loss [69, 70].

10.5 Mechanisms of acupuncture treatment on obesity linked PCO

Acupuncture therapy is believed to regulate the hypothalamic–pituitary axis and neuroendocrine. It modulates energy metabolism and eating habits, gastrointestinal tract (GIT) secretions, GI hormones and classic hormones secretions, and nervous system functions [71]. The hypothalamus arcuate nucleus (ARC) contains two microcircuits, agouti-related peptide (AgRP) and neuropeptide-(NPY), which are important for energy balance and have an antagonistic role’. Its signals to stimulate hunger and food intake, and the POMC (pro-opiomelanocortin) signals reduce food intake and satiety. ARC is involved in the central regulation of body weight, energy expenditure, and selected food intake, stimulation of this area alters food intake [72]. Animal model study reveals that EAT reduces food intake; and weight loss and increases the peptide α-MSH and mRNA expression in ARH. Furthermore, EAT significantly reduces the energy intake and body weight in 14-week high-fat diet-fed rats and upregulates the amphetamine-regulated transcript (CART) and cocaine in ARC. It indicates that the valuable effects of EAT act through ARC [73, 74].

Furthermore, specially obesity-linked areas are the Satiety center” in the ventromedial hypothalamus (VMH) and “the feeding center” in the lateral hypothalamus (LH) [75]. Stimulation of these LH centers through EA reduces the excitation of LH, and inhibits hyperorexia-obesity and regulates the catecholamine, ATPase, 5-hydroxytryptamine (5HT) levels in LH [76, 77]. In 1999, Su et al. reported that stimulation of the satiety center, tends to prevent obesity. Through EAT stimulation of the VMH center the levels of dopamine (DA), tyrosine (Tyr), and tryptophan (Typ) ratio were elevated, increasing the excitability of VMH [78].

Research studies believe that normal acupuncture or EAT stimulation increases the spontaneous discharges of nerve cells of the paraventricular nucleus (PVN) important area for feeding and drinking and decreases the activity of the perifornical nucleus (PeF) The PeF nucleus is important for wakefulness and sleep [79, 80]. Stimulation of the most important acupuncture point in the Stomach 36 (ST36, EA zusanli) upregulates the proopiomelanocortin (POMC) in neurons of the Nucleus Tractus solitarius (NTS) and hypoglossal nucleus (HN) preventing food intake and leading to weight loss (Figure 5) [81].

Figure 5.

Neuroendocrine regulation under the effectiveness of acupuncture treatment in the animal model studies. The arcuate nucleus of the hypothalamus (ARH), Ventrosomedial hypothalamus (DMH) lateral hypothalamic area (LHA), paraventricular nucleus (PVN), perifornical nucleus (PeF) Ref [82].

10.6 Mechanisms of acupuncture treatment on insulin resistance with PCO

Most PCO women are insulin-resistant, particularly obese females [83]. When cells do not respond to insulin, they cannot easily uptake glucose from the blood. It leads to hyperglycemia and excess insulin in the blood (hyperinsulinemia), both of which induce reproductive failure and PCO [84]. Many clinical studies have proved that EAT has beneficial effects on insulin resistance in PCOS patients, and EAT inhibited the insulin resistance of PCO females via activation of the IRS-1/PI3K/GLUT4 pathway [25, 26]. In 2020, Peng Y et al. [7]. reported that in a PCO rat model study, EAT activates the AMPK pathway to suppress SREBP-1 expression, ultimately, inactivating insulin resistance, oxidative stress, and mitochondrial dysfunction (Figure 6).

Figure 6.

The effects of EAT on the insulin pathway. EAT upregulates the insulin receptor substrates-1/PI3K/GLUT4 pathway and increases the GLUT4 expression or triggers the adenosine monophosphate-activated protein kinase (AMPK) pathway or stops the PI3K/AKT/mTOR pathway. It also decreases the levels of tumor necrosis factor-α and interleukin-6 both are involved in insulin resistance Ye et al. [85].

The present book chapter summarizes various clinical studies of women with PCOs and PCO animal model studies. Further, it explains the mechanisms of acupuncture and EAT on PCOS-related key symptoms of hyperandrogenism, insulin resistance, ovulatory dysfunction, obesity, and negative emotion. Further, the main acupoints were used for treating women with PCOS, and animals were also described in this chapter.

References

  1. 1. Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990-2010: A systematic analysis for the global burden of disease study 2010. Lancet. 2012;380(9859):2163-2196
  2. 2. Escobar-Morreale HF. Polycystic ovary syndrome: Definition, aetiology, diagnosis and treatment. Nature Reviews. Endocrinology. 2018;14:270-284
  3. 3. Zhang J, Xin X, Zhang H, Zhu Y, Ye Y, Li D. The efficacy of Chinese herbal medicine in animal models of polycystic ovary syndrome: A systematic review and meta-analysis. Evidence-based Complementary and Alternative Medicine. 2022;2022:4892215
  4. 4. Chen Z, Liu L, Xi X, Burn M, Karakaya C, Kallen AN. Aberrant H19 expression disrupts ovarian Cyp17 and testosterone production and is associated with polycystic ovary syndrome in women. Reproductive Sciences. 2022;29:1357-1367
  5. 5. Palomba S, Piltonen TT, Giudice LC. Endometrial function in women with polycystic ovary syndrome: A comprehensive review. Human Reproduction Update. 2021;27:584-618
  6. 6. Calcaterra V, Verduci E, Cena H, Magenes VC, Todisco CF, Tenuta E, et al. Polycystic ovary syndrome in insulin-resistant adolescents with obesity: The role of nutrition therapy and food supplements as a strategy to protect fertility. Nutrients. 2021;13:1848
  7. 7. Peng Y, Guo L, Gu A, Shi B, Ren Y, Cong J, et al. Electroacupuncture alleviates polycystic ovary syndrome-like symptoms through improving insulin resistance, mitochondrial dysfunction, and endoplasmic reticulum stress via enhancing autophagy in rats. Molecular Medicine. 2020;26:73
  8. 8. Bednarska S, Siejka A. The pathogenesis and treatment of polycystic ovary syndrome: What’s new? Advances in Clinical and Experimental Medicine. 2017;26:359-367. DOI: 10.17219/acem/59380
  9. 9. Liu H-Y, Liu J-Q , Mai Z-X, Zeng Y-T. A subpathway-based method of drug reposition for polycystic ovary syndrome. Reproductive Sciences. 2014;22:423-430
  10. 10. Li Y, Zheng Q , Sun D, Cui X, Chen S, Bulbul A, et al. Dehydroepiandrosterone stimulates inflammation and impairs ovarian functions of polycystic ovary syndrome. Journal of Cellular Physiology. 2018;234:7435-7447. DOI: 10.1002/jcp.27501
  11. 11. Vaklavas C, Roberts BS, Varley KE, Lin NU, Liu MC, Rugo HS, et al. TBCRC 002: A phase II, randomized, open-label trial of preoperative letrozole with or without bevacizumab in postmenopausal women with newly diagnosed stage 2/3 hormone receptor-positive and HER2-negative breast cancer. Breast Cancer Research. 2020;22:22
  12. 12. Wang R, Li W, Bordewijk EM, Legro RS, Zhang H, Wu X, et al. First-line ovulation induction for polycystic ovary syndrome: An individual participant data meta-analysis. Human Reproduction Update. 2019;25:717-732
  13. 13. Balen AH, Morley LC, Misso M, Franks S, Legro RS, Wijeyaratne CN, et al. The management of anovulatory infertility in women with polycystic ovary syndrome: An analysis of the evidence to support the development of global WHO guidance. Human Reproduction Update. 2016;22:687-708
  14. 14. Jia L-Y, Feng J-X, Li J-L, Liu F-Y, Xie L-Z, Luo S-J, et al. The complementary and alternative medicine for polycystic ovary syndrome: A review of clinical application and mechanism. Evidence-Based Complementary Alternatve Medicine. 2021;2021:1-12
  15. 15. Rudnicka E, Suchta K, Grymowicz M, Calik-Ksepka A, Smolarczyk K, Duszewska A, et al. Chronic low grade inflammation in pathogenesis of PCOS. International Journal of Molecular Sciences. 2021;22:3789. DOI: 10.3390/ijms22073789
  16. 16. Lu L, Zhang Y, Tang X, Ge S, Wen H, Zeng J, et al. Evidence on acupuncture therapies is underused in clinical practice and health policy. BMJ. 2022;376:e067475
  17. 17. Zhang B, Shi H, Cao S, Xie L, Ren P, Wang J, et al. Revealing the magic of acupuncture based on biological mechanisms: A literature review. Bioscience Trends. 2022;16:73-90
  18. 18. Quiroz-González S, Torres- Castillo S, López-Gómez RE, Jiménez EI. Acupuncture points and their relationship with multireceptive fields of neurons. Journal of Acupuncture Meridian Studies. Apr 2017;10(2):81-89
  19. 19. Vickers AJ, Vertosick EA, Lewith G, MacPherson H, Foster NE, Sherman KJ, et al. Acupuncture for chronic pain: update of an individual patient data meta-analysis. Journal of Pain. 2018;19(5):455-474
  20. 20. Kong J, Ma L, Gollub RL, Wei J, Yang X, Li D, et al. A pilot study of functional magnetic resonance imaging of the brain during manual and Electroacupuncture stimulation of acupuncture point (LI-4 Hegu) in Normal subjects reveals differential brain activation between methods the. Journal of Alternative and Complementary Medicine. 2004;8(4)
  21. 21. Dhond RP, Kettner N, Napadow V. Neuroimaging acupuncture effects in the human brain. Journal of Alternative and Complementary Medicine. 2007;13(6):603-616
  22. 22. Bianchi M, Jotti E, Sacerdote P, Panerai AE. Traditional acupuncture increases the content of beta-endorphin in immune cells and influences mitogen induced proliferation. American Journal of China Medicine. 1991;19:101-104
  23. 23. Wu J, Chen D, Liu N. Effectiveness of acupuncture in polycystic ovary syndrome: A systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore). 2020;99:e20441
  24. 24. Kokosar M, Benrick A, Perfilyev A, Nilsson E, Kallman T, Ohlsson C, et al. A single bout of electroacupuncture remodels epigenetic and transcriptional changes in adipose tissue in polycystic ovary syndrome. Scientific Reports. 2018;8:1878
  25. 25. Li J, Wu W, Stener-Victorin E, Ng EHY, Li RHW, Li M, et al. A prospective pilot study of the effect of acupuncture on insulin sensitivity in women with polycystic ovary syndrome and insulin resistance. Acupuncture in Medicine. 2020;38:310-318
  26. 26. Xiang S, Xia MF, Song JY, Liu DQ , Lian F. Effect of electro-acupuncture on expression of IRS-1/PI3K/GLUT4 pathway in ovarian granulosa cells of infertile patients with polycystic ovary syndrome-insulin resistance of phlegm-dampness syndrome. Chinese Journal of Integrative Medicine. 2021;27:330-335
  27. 27. Pan W, Li FX, Wang Q , Huang ZQ , Yan YM, Zhao L, et al. A randomized sham-controlled trial of manual acupuncture for infertile women with polycystic ovary syndrome. Integrative Medical Research. 2022;11:100830
  28. 28. Johansson J, Redman L, Veldhuis PP, Sazonova A, Labrie F, Holm G, et al. Acupuncture for ovulation induction in polycystic ovary syndrome: A randomized controlled trial. American Journal of Physiology. Endocrinology and Metabolism. 2013;304(9):E934-E943
  29. 29. Zheng YH, Wang XH, Lai MH, Yao H, Liu H, Ma HX. Effectiveness of abdominal acupuncture for patients with obesity-type polycystic ovary syndrome: A randomized controlled trial. Journal of Alternative and Complementary Medicine. 2013;19(9):740-745
  30. 30. Li Y, Zhang L, Gao J, Yan J, Feng X, He X, et al. Effect of acupuncture on polycystic ovary syndrome in animal models: A systematic review. Evidence-based Complementary and Alternative Medicine. 2021;2021:5595478
  31. 31. Stener-Victorin E, Holm G, Janson PO, Gustafson D, Waern M. Acupuncture and physical exercise for affective symptoms and health-related quality of life in polycystic ovary syndrome: Secondary analysis from a randomized controlled trial. BMC Complementary and Alternative Medicine. 2013;13:131
  32. 32. Maliqueo M, Benrick A, Alvi A, Johansson J, Sun M, Labrie F, et al. Circulating gonadotropins and ovarian adiponectin system are modulated by acupuncture independently of sex steroid or beta-adrenergic action in a female hyperandrogenic rat model of polycystic ovary syndrome. Molecular and Cellular Endocrinology. 2015;412:159
  33. 33. Jedel E, Labrie F, Oden A, Holm G, Nilsson L, Janson PO, et al. Impact of electro-acupuncture and physical exercise on hyperandrogenism and oligo/amenorrhea in women with polycystic ovary syndrome: A randomized controlled trial. American Journal of Physiology. Endocrinology and Metabolism. 2011a;300(1):E37-E45
  34. 34. Lim DC, Chen W, Cheng LN, Xue CC, Wong FW, O’Sullivan AJ, et al. Acupuncture for polycystic ovarian syndrome. Cochrane Database of Systematic Reviews. 2011;8:CD007689
  35. 35. Chen J, Feng S, Zeng J, Wu X, Yang M, Tang H, et al. Effectiveness of electroacupuncture for polycystic ovary syndrome: Study protocol for a randomized controlled trial. Trials. 2016;17(1):256
  36. 36. Ramadoss M, Ramanathan G. Effect of electroacupuncture on anovulation, epinephrine and norepinephrine levels in drug induced polycystic ovary rats. International Journal of Science and Research (IJSR). 2017;6(1)
  37. 37. Stener-Victorin E, Lundeberg T, Waldenstrom U, et al. Effects of electro-acupuncture on nerve growth factor and ovarian morphology in rats with experimentally induced polycystic ovaries. Biology of Reproduction. 2000;63(5):1497-1503
  38. 38. Brawer JR, Munoz M, Farookhi R. Development of the polycystic ovarian condition (PCO) in the estradiol valerate treated rat. Biology of Reproduction. 1986;35(3):647-655
  39. 39. Dumesic DA, Hoyos LR, Chazenbalk GD, Naik R, Padmanabhan V, Abbott DH. Mechanisms of intergenerational transmission of polycystic ovary syndrome. Reproduction. 2020;159:R1-R13
  40. 40. Budihastuti UR, Melinawati E, Sulistyowati S, Nurwati I. Electro-acupuncture effect on polycystic ovary syndrome to improve oocytes’ growth. Medical Acupuncture. 2019;31:379-383
  41. 41. Rashidi BH, Tehrani ES, Hamedani NA, Pirzadeh L. Effects of acupuncture on the outcome of in vitro fertilisation and intracytoplasmic sperm injection in women with polycystic ovarian syndrome. Acupuncture in Medicine. 2013;31:151-156. DOI: 10.1136/acupmed-2012-010198
  42. 42. Chen X, Tang H, Liang Y, Wu P, Xie L, Ding Y, et al. Acupuncture regulates the autophagy of ovarian granulosa cells in polycystic ovarian syndrome ovulation disorder by inhibiting the PI3K/AKT/mTOR pathway through LncMEG3. Biomedicine & Pharmacotherapy. 2021;144:112288
  43. 43. Shi Y, Li L, Zhou J, Sun J, Chen L, Zhao J, et al. Efficacy of electroacupuncture in regulating the imbalance of AMH and FSH to improve follicle development and hyperandrogenism in PCOS rats. Biomedicine & Pharmacotherapy. 2019;113:108687
  44. 44. Tong X, Liu Y, Xu X, Shi J, Hu W, Ma T, et al. Ovarian innervation coupling with vascularity: The role of electro-acupuncture in follicular maturation in a rat model of polycystic ovary syndrome. Frontiers in Physiology. 2020;11:474
  45. 45. Krishnan A, Muthusami S. Hormonal alterations in PCOS and its influence on bone metabolism. The Journal of Endocrinology. 2017;232:R99-R113
  46. 46. Wang Z, Yang L, Dong H, Dong H, Cheng L, Yi P, et al. Effect of electroacupuncture on the kisspeptin system in a pubertal rat model of polycystic ovary syndrome. Acupuncture in Medicine. 2021;39:491-500
  47. 47. Ramadoss M, Ramanathan G, Subbiah AJ, Natrajan C. Heart rate changes in electroacupuncture treated polycystic ovary in rats. Journal of Clinical and Diagnostic Research. 2016;10(3):CF01-3
  48. 48. Task Force of the European Society of Cardiology and the North American Society of Pacing and Electrophysiology. Heart rate variability: Standards of measurement, physiological interpretation and clinical use. Circulation. 1996;93(5):1043-1065
  49. 49. Lara HE, Dissen GA, Leyton V, Paredes A, Fuenzalida H, Fiedler JL, et al. An increased intraovarian synthesis of nerve growth factor and its low affinity receptor is a principal component of steroid-induced polycystic ovary in the rat. Endocrinology. 2000;141(3):1059-1072
  50. 50. Chintala KK, Krishna BH, N MR. Heart rate variability in overweight health care students: Correlation with visceral fat. Journal of Clinical and Diagnostic Research. 2015;9(1):CC06-CC08
  51. 51. Schmidt J, Landin-Wilhelmsen K, Brännström M, Dahlgren E. Cardiovascular disease and risk factors in PCOS women of postmenopausal age: A 21-year controlled follow-up study. The Journal of Clinical Endocrinology and Metabolism. 2011;96(12):3794-3803
  52. 52. Stener-Victorin E, Jedel E, Janson PO, Sverrisdottir YB. Low-frequency electroacupuncture and physical exercise decrease high muscle sympathetic nerve activity in polycystic ovary syndrome. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2009;297(2):R387-R395
  53. 53. Stener-Victorin E, Lundeberg T, Cajander S, et al. Steroid-induced polycystic ovaries in rats: Effect of electro-acupuncture on concentrations of endothelin-1 and nerve growth factor (NGF), and expression of NGF mRNA in the ovaries, the adrenal glands, and the central nervous system. Reproductive Biology and Endocrinology. 2003;1:33
  54. 54. Mannerås L, Cajander S, Lönn M, Stener-Victorin E. Acupuncture and exercise restore adipose tissue expression of sympathetic markers and improve ovarian morphology in rats with dihydrotestosterone-induced PCOS. American Journal of Physiology. 2009;296(4):R1124-R1131
  55. 55. Manni L, Lundeberg T, Holmäng A, Aloe L, Stener-Victorin E. Effect of electro-acupuncture on ovarian expression of α(1)- and β(2)-adrenoceptors, and p75 neurotrophin receptors in rats with steroid-induced polycystic ovaries. Reproductive Biology and Endocrinology. 2005;3:article 21
  56. 56. Stener-Victorin E, Deng Q. Epigenetic inheritance of polycystic ovary syndrome – challenges and opportunities for treatment. Nature Reviews. Endocrinology. 2021;17:521-533
  57. 57. Feng Y, Johansson J, Shao R, Manneras-Holm L, Billig H, Stener-Victorin E. Electrical and manual acupuncture stimulation affect oestrous cyclicity and neuroendocrine function in an 5alpha-dihydrotestosterone-induced rat polycystic ovary syndrome model. Experimental Physiology. 2012;97:651-662
  58. 58. Zhao QY, Sun Y, Zhou J, Gao YL, Ma GZ, Hu ZH, et al. Effectiveness of herb-partitioned moxibustion combined with electroacupuncture on polycystic ovary syndrome in patients with symptom pattern of kidney deficiency and phlegm-dampne. Journal of Traditional Chinese Medicine. 2021;41:985-993
  59. 59. Sun J, Jin C, Wu H, Zhao J, Cui Y, Liu H, et al. Effects of electro-acupuncture on ovarian P450arom, P450c17alpha and mRNA expression induced by letrozole in PCOS rats. PLoS One. 2013;8:e79382
  60. 60. Xu G, Zhang A, Liu J, Wang X, Feng J, Chen Y. Effects of electroacupuncture on ovarian expression of the androgen receptor and connexin 43 in rats with letrozole-induced polycystic ovaries. Evidence-based Complementary and Alternative Medicine. 2020;2020:3608062
  61. 61. Balen AH, Conway GS, Kaltsas G, et al. Polycystic ovary syndrome: The spectrum of the disorder in 1741 patients. Human Reproduction. 1995;10:2107-2111
  62. 62. Barber TM, McCarthy MI, Wass JA, Franks S. Obesity and polycystic ovary syndrome. Clinical Endocrinology. 2006;65:137-145
  63. 63. Legro RS. The genetics of obesity. Lessons for polycystic ovary syndrome. Annals of the New York Academy of Sciences. 2000;900:193-202
  64. 64. Kiddy DS, Hamilton-Fairley D, Bush A, et al. Improvement in endocrine and ovarian function during dietary treatment of obese women with polycystic ovary syndrome. Clinical Endocrinology. 1992;36:105-111
  65. 65. Holte J, Bergh T, Berne C, Wide L, Lithell H. Restored insulin sensitivity but persistently increased early insulin secretion after weight loss in obese women with polycystic ovary syndrome. The Journal of Clinical Endocrinology and Metabolism. 1995;80:2586-2593
  66. 66. The Cooperative Group Will Be Evaluated after the Listing of Sibutramine Center. Clinical curative effect and safety research in treatment of the simple obesity with sibutramine hydrochloride capsule. Chinese Journal of Clinical Pharmacy. 2004;15:17-20
  67. 67. Wu XK, Stener-Victorin E, Kuang HY, et al. Effect of acupuncture and clomiphene in Chinese women with polycystic ovary syndrome: A randomized clinical trial. Journal of the American Medical Association. 2017;317(24):2502-2514
  68. 68. Liu Z, Yan S, Wu J, et al. Acupuncture for chronic severe functional constipation: A randomized trial. Annals of Internal Medicine. 2016;165(11):761-769
  69. 69. Richards D, Marley J. Stimulation of auricular acupuncture points in weight loss. Australian Family Physician. 1998;27(Suppl 2):S73-S77
  70. 70. Mannerås L, Jonsdottir IH, Holmäng A, et al. Low-frequency electroacupuncture and physical exercise improve metabolic disturbances and modulate gene expression in adipose tissue in rats with dihydrotestosterone-induced polycystic ovary syndrome. Endocrinology. 2008;149:3559-3568
  71. 71. Hwang SC, Wong EK, Son Y, et al. Neural substrates, experimental evidence, and functional hypothesis of acupuncture mechanisms. Acta Neurologica Scandinavica. 2006;113(6):370-377
  72. 72. Joly-Amado A, Cansell C, Denis RG, Delbes AS, Castel J, Martinez S, et al. The hypothalamic arcuate nucleus and the control of peripheral substrates. Best Practice & Research. Clinical Endocrinology & Metabolism. 2014;28(5):725-737
  73. 73. Wang F, Tian DR, Tso P, Han JS. Arcuate nucleus of hypothalamus is involved in mediating the satiety effect of electroacupuncture in obese rats. Peptides. 2011;32(12):2394-2399
  74. 74. Tian D-R, Li X-D, Wang F, et al. Up-regulation of the expression of cocaine and amphetamine-regulated transcript peptide by electroacupuncture in the arcuate nucleus of diet-induced obese rats. Neuroscience Letters. 2005;383(1-2):17-21
  75. 75. Elmquist JK, Elias CF, Saper CB. From lesions to leptin: Hypothalamic control of food intake and body weight. Neuron. 1999;22:221-232
  76. 76. Liu ZC, Sun MF, Han Y, et al. Effect of acupuncture on level of monoamines and activity of adenosine triphosphatase in lateral hypothalamic area og obese rats. Chinese Journal of Integrated Traditional and Western Medicine. 2000;7:521-523
  77. 77. Zhao M et al. Effect of acupuncture on feeding center of hypothalamus in experimental fat rats. Chinese Acupuncture & Moxibustion. 2001;5:49-51
  78. 78. Liu ZC, Sun FM, Su J, et al. Study on action of acupuncture on ventromedial nucleus of hypothalamus in obese rats. Journal of Traditional Chinese Medicine. 2001;3:220-224
  79. 79. Liu ZC, Sun MF, Zhao M, et al. Effect of acupuncture on paraventricular nucleus of obese rats. Journal of Traditional Chinese Medicine. 2003;7:1031-1033, 1059
  80. 80. Sun Z, Zhang ZC, Ma L, Liu ZC. Effect of acupuncture on expression of hypothalamic neuropeptide Y gene in obese rats. Chinese Journal of Tissue Engineering Research. 2006;15:135-138
  81. 81. Ji B, Hu J, Ma S. Effects of electroacupuncture Zusanli (ST36) on food intake and expression of POMC and TRPV1 through afferents-medulla pathway in obese prone rats. Peptides. 2013;40:188-194. DOI: 10.1016/j.peptides.2012.10.009
  82. 82. Zhang K, Zhou S, Wang C, Xu H, Zhang L. Acupuncture on obesity: Clinical evidence and possible neuroendocrine mechanisms. Evidence-based Complementary and Alternative Medicine. 2018;2018:6409389
  83. 83. Diamanti-Kandarakis E, Dunaif A. Insulin resistance and the polycystic ovary syndrome revisited: An update on mechanisms and implications. Endocrine Reviews. 2012;33:981-1030
  84. 84. Moghetti P, Tosi F. Insulin resistance and PCOS: Chicken or egg? Journal of Endocrinological Investigation. 2021;44:233-244
  85. 85. Ye Y, Zhou CC, Hu HQ , Fukuzawa I, Zhang HL. Underlying mechanisms of acupuncture therapy on polycystic ovary syndrome: Evidences from animal and clinical studies. Frontiers in Endocrinology (Lausanne). 2022;13:1035929

Written By

Gunasekaran Ramanathan

Submitted: 24 August 2023 Reviewed: 20 October 2023 Published: 19 December 2023