1
|
Su FZ, Bai CX, Zhang WS, Liu M, Li B, Sun MH, He YJ, Zeng YN, Sun YP, Yang BY, Kuang HX, Wang QH. Lipid-lowering effects of bile Arisaema polysaccharides on high-fat diet-induced hyperlipidemia: An integrated analysis of metabolomics, lipidomics and microbiome. Int J Biol Macromol 2025; 311:143932. [PMID: 40348213 DOI: 10.1016/j.ijbiomac.2025.143932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025]
Abstract
Bile Arisaema, a traditional Chinese medicine, has been previously identified by our team to possess antipyretic properties attributed to its polysaccharide component. Recently, we have confirmed that bile Arisaema played a significantly lipid-lowering effect on hyperlipidemia rats. Building upon this discovery, the present study aimed to investigate the unexplored hypolipidemic potential of its polysaccharide component and elucidate the underlying mechanisms. A soluble polysaccharide fraction devoid of free proteins, named BAPs, was extracted from bile Arisaema using a combination of hot water extraction, alcohol precipitation, and the Sevage method. The structural characteristics of BAPs were preliminarily elucidated through monosaccharide composition analysis (mainly composed of glucose), molecular weight distribution (38.74 kDa and 2.87 kDa), and glycosyl linkage analysis via methylation. The results of animal experiment demonstrated that oral administration of BAPs (400 mg/kg/day) for four weeks significantly improved abnormal serum lipid levels, hepatic function and histopathological injury on high-fat diet-induced hyperlipidemia rats. Mechanistically, the results of high throughput sequencing indicated that BAPs intake markedly altered the hepatic and fecal metabolome and lipidome, while also modulating gut microbiota composition and improving intestinal barrier integrity. Spearman's correlation analysis unveiled closely associations between the altered microbes, lipids, metabolites and serum biochemical indicators. Western blotting and qRT-PCR analyses further confirmed that these metabolic improvements were mediated by the regulation of key genes involved in lipid metabolism. Collectively, this study demonstrated that BAP supplementation effectively improved serum lipid profiles in hyperlipidemia rats by modulating metabolic disorders and restoring gut homeostasis.
Collapse
Affiliation(s)
- Fa-Zhi Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Chen-Xi Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Wen-Sen Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Meng Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Biao Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Ming-Hao Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Yu-Jia He
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Yuan-Ning Zeng
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yan-Ping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Bing-You Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China
| | - Hai-Xue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China.
| | - Qiu-Hong Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin 150040, China; School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
2
|
Racine C, Fraissinet F, Tolu S, Pereira T, Gil S, Badel A, Bailbé D, Fève B, Movassat J, Cate R, di Clemente N. A blocking antibody against anti-Müllerian hormone restores ovulation and normal androgen levels in a spontaneous rat model of polycystic ovary syndrome. EBioMedicine 2025; 115:105716. [PMID: 40252252 PMCID: PMC12032919 DOI: 10.1016/j.ebiom.2025.105716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Polycystic Ovary Syndrome (PCOS), the leading cause of infertility worldwide, is characterised by oligo-anovulation, hyperandrogenism, polycystic ovarian morphology and high Anti-Müllerian hormone (AMH) levels, associated with severe metabolic disturbances. However, the role of AMH in the physiopathology of this syndrome remains poorly understood and strategies to block its effects have never been investigated in animal models of PCOS. METHODS We used Western-blotting, ELISA and gene reporter approaches to evaluate the blocking efficacy, interspecificity and mechanism of action of an antibody against human AMH, Mab22A2. Then, we investigated the ability of a rat version of Mab22A2, rMab22A2, to alleviate reproductive dysfunction in Goto-Kakizaki (GK) rats, which spontaneously exhibit all the features of women with PCOS. FINDINGS We showed that Mab22A2 was interspecific, did not prevent AMH from binding to its receptor and was able to block the effects of AMH in gonadal cell lines. In addition, treatment of anovulatory GK rats with rMab22A2 reduced their bioavailable serum AMH levels and normalised their androgen concentrations. Finally, this treatment also induced ovulation in 84% of the rats and resulted in 66% of pregnancies. INTERPRETATION Our results show that AMH is a major driver of reproductive and hormonal dysfunction in PCOS and provide proof of concept that a blocking antibody against AMH can reverse the major reproductive dysfunction observed in PCOS, opening up promising avenues for the treatment of patients with PCOS. FUNDING Inserm, Sorbonne University, Inserm Transfert, the French Endocrine Society and the Medical Research Foundation (grant agreement n°EQU201903007868).
Collapse
Affiliation(s)
- Chrystèle Racine
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Université Paris Cité, Paris 75013, France
| | - François Fraissinet
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | - Stefania Tolu
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Tony Pereira
- Department of General Biochemistry, Rouen University Hospital, Rouen 76000, France
| | | | - Anne Badel
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Danielle Bailbé
- Université Paris Cité, BFA, UMR8251, CNRS, Paris 75013, France
| | - Bruno Fève
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France; Department of Endocrinology, Hôpital Saint-Antoine, APHP, Paris 75012, France
| | | | - Richard Cate
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Nathalie di Clemente
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), UMR_S938, Paris 75012, France; Institut Hospitalo-Universitaire ICAN, Paris 75013, France.
| |
Collapse
|
3
|
Tong C, Wu Y, Zhuang Z, Yu Y. A diagnostic model for polycystic ovary syndrome based on machine learning. Sci Rep 2025; 15:9821. [PMID: 40119083 PMCID: PMC11928512 DOI: 10.1038/s41598-025-92630-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
Diagnosis of polycystic ovary syndrome remains a challenge. In this study, we propose constructing a diagnostic model of polycystic ovary syndrome by combining anti-Müllerian hormone with steroid hormones and oestrogens, with the aim of providing more bases and auxiliary means for the diagnosis of this disease. 1. Eighty-four samples from patients who were diagnosed with polycystic ovary syndrome at the First Affiliated Hospital of Zhejiang Chinese Medical University from May 2023 to November 2023 were collected as the case group, and 75 samples from the healthy population of the Health Screening Centre of the First Affiliated Hospital of Zhejiang Chinese Medical University during the same period were collected as the control group. 2. General information (including age, BMI, family history, medication history, etc.) and sex hormone data (including luteinising hormone, follicle stimulating hormone, prolactin, estradiol, testosterone, etc.) were collected from all study subjects. AMH and steroid hormone tests were performed on serum collected from all study subjects. 3. The data of 10 case groups and 10 control groups were randomly selected as validation set data, and the rest of the data were included in the model construction. The acquired data were screened for variables, a classification model based on a machine learning algorithm was constructed, and the constructed model was evaluated and validated for diagnostic efficacy. Ultimately, a total of 8 variables were screened and included in the subsequent model construction, namely LH, LH/FSH, E2, PRL, T, AMH, AD, and COR, with AMH having the highest diagnostic potential among all the variables included in the model. A total of five machine learning models were constructed, the logistic classification model has the best overall performance, and the support vector machine has the weakest overall performance. The validation set has an AUC of 0.86 for the model. In this study, five classification models based on machine learning algorithms were successfully constructed. Combining the evaluation metrics of each model performance, we concluded that the logistic classification model had the best performance capability in our study. However, since this study is a single-center small sample size study, some metabolic features of PCOS may be overlooked, and, as the validation set data in this study come from the same center as the modelling data, the validation results may have several limitations, so it is still necessary to expand the sample size and collect multicenter data to establish an external validation dataset to further improve the study.
Collapse
Affiliation(s)
- Cheng Tong
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Yue Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China
| | - Zhenchao Zhuang
- Adicon Clinical Laboratories, Hangzhou, 310023, Zhejiang, China.
| | - Ying Yu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
4
|
Yang W, Qiu J, Zi J, Li Y, Li J, Guo M, Zhou Y, Yang X, Lai Y. Effect of Rhei Radix Et Rhizome on treatment of polycystic ovary syndrome by regulating PI3K/AKT pathway and targeting EGFR/ALB in rats. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119020. [PMID: 39491761 DOI: 10.1016/j.jep.2024.119020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Abnormal endocrine metabolism caused by polycystic ovary syndrome (PCOS) poses a serious risk to reproductive health in females. According to Traditional Chinese Medicine (TCM) theories, the leading causes of PCOS include turbid phlegm, blood stasis and stagnation of liver Qi. Rhei Radix Et Rhizome is widely used in TCM to attack stagnation, clear damp heat, relieve fire. Rhei Radix Et Rhizome is an important part of the TCM formulas for the treatment of PCOS, which has a long history of medicinal use. However, the specific effect and mechanisms of Rhei Radix Et Rhizome on PCOS have yet to be elucidated. AIM OF THE STUDY The object of this study aimed to investigate the effect and its pharmacological mechanism of Rhei Radix Et Rhizome on the treatment of polycystic ovary syndrome. METHODS PCOS was induced in female Sprague Dawley (SD) rats by administering letrozole (1 mg/kg, per orally, p.o.) for 21 days, then treated with Rhei Radix Et Rhizome at doses of 0.6 g/kg or 1.2 g/kg. Rats weight, blood glucose and estrus period are measured, and serum hormone include free testosterone (T), luteinizing hormone (LH), follicle-stimulating hormone (FSH) and ovarian lesions were observed to determine the effects of Rhei Radix Et Rhizome. Network pharmacology and molecular docking predicted the targets of Rhei Radix Et Rhizome on PCOS. Epidermal growth factor receptor (EGFR), albumin (ALB), PI3K and P-AKT/AKT protein expression levels in ovarian tissues were assessed by Western blot. RESULTS Rhei Radix Et Rhizome reduce abnormal weight and fasting blood glucose induced by letrozole (n = 5, p < 0.01), and improve the disturbed estrus cycle, reduce T, LH levels and LH/FSH ratio of PCOS rats (n = 4, p < 0.01). In addition, it alleviates the polycystic changes of ovaries in PCOS rats and reduces ovarian histopathological damage (n = 4, p < 0.01). Additionally, the core active components of Rhei Radix Et Rhizome for PCOS include Sennoside D_qt, Procyanidin B-5,3'-O-gallate, and Mutatochrome, which strongly bind to core therapeutic targets ALB and EGFR. Furthermore, the treatment reduces the increase of EGFR and ALB induced by letrozole (n = 4, p < 0.01). KEGG pathway enrichment analysis highlights endocrine resistance and prolactin signaling pathway, in both of which the PI3K/AKT pathway plays a crucial role. Our results show Rhei Radix Et Rhizome rescue the abnormal expression of PI3K/AKT pathway in PCOS rats (n = 4, p < 0.01). However, no significant dose-dependent relationship was observed in the tested dose range for the above experiments. CONCLUSION These findings suggest that Rhei Radix Et Rhizome can regulate the PI3K/AKT pathway and target EGFR and ALB to treat polycystic ovary syndrome in rats. This study provides a scientific basis for the use of Rhei Radix Et Rhizome in the treatment of PCOS and highlights its potential mechanism through modulation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Wanqi Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, Yunnan Province, PR China.
| | - Jishuang Qiu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Jiangli Zi
- College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Yang Li
- College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Jiao Li
- College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Meixian Guo
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, Yunnan Province, PR China
| | - Yanru Zhou
- College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Xiaotong Yang
- College of Pharmacy, Dali University, Dali, Yunnan, PR China
| | - Yong Lai
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, PR China; College of Pharmacy, Dali University, Dali, Yunnan, PR China; National-Local Joint Engineering Research Center of Entomoceutics, Dali University, Dali, Yunnan Province, PR China.
| |
Collapse
|
5
|
Javed M, Suramya, Mangla A, Jindal G, Bhutto HN, Shahid S, Kumar S, Raisuddin S. Bisphenol A-induced polycystic ovary syndrome (PCOS) with hormonal and metabolic implications in rats. Reprod Toxicol 2025; 131:108750. [PMID: 39549768 DOI: 10.1016/j.reprotox.2024.108750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
There is a rising incidence of polycystic ovary syndrome (PCOS) cases worldwide in women of reproductive age due to environmental factors. We evaluated the effect of an environmental estrogen, bisphenol A (BPA) for its reprotoxicity regarding the induction of PCOS in rats and also assessed its hormonal and metabolic implications. There was 66.6 % and 50 % disorder, in the estrus cycle at low (50 µg/kg) and high (500 µg/kg) doses of BPA, respectively. While animals treated with the positive control (dehydroepiandrosterone, DHEA at 6 mg/100 g) caused 100 % disorder. Cystic and atretic follicles along with two corpora lutea were found in the low dose group. However, no corpus luteum was found in the high dose group. Furthermore, hyperplasia and hypertrophy were found in the myometrium, endometrium, and luminal epithelium of the uterus of the low dose and DHEA groups. Additionally, 17β estradiol, progesterone, DHEA, androstenedione, testosterone, dihydrotestosterone (DHT), dehydroepiandrosterone sulphate (DHEAS), antimullerian hormone (AMH), ratio of LH/FSH and testosterone/DHT were increased significantly (P < 0.01) in BPA groups. A significantly higher TSH (P < 0.01) indicates hypothyroidism. Furthermore, hyperglycemia, hyperinsulinemia, HOMA-IR, and HOMAβ indicate insulin resistance in the low-dose group. Thus, the low dose of BPA was found to be more potent as compared to the higher dose in defining the hyperandrogenic state. Our study revealed that BPA may not only be a causative factor in the induction of PCOS but also has metabolic implications bearing on its estrogenic nature.
Collapse
Affiliation(s)
- Mehjbeen Javed
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suramya
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Anuradha Mangla
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Garima Jindal
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Humaira Naaz Bhutto
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Shaesta Shahid
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suraj Kumar
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Sheikh Raisuddin
- Molecular Toxicology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
6
|
Hu R, Huang Y, Liu Z, Dong H, Ma W, Song K, Xu X, Wu X, Geng Y, Li F, Zhang M, Song Y. Characteristics of polycystic ovary syndrome rat models induced by letrozole, testosterone propionate and high-fat diets. Reprod Biomed Online 2025; 50:104296. [PMID: 39626468 DOI: 10.1016/j.rbmo.2024.104296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 01/29/2025]
Abstract
RESEARCH QUESTION What are the long-term effects of different models of polycystic ovary syndrome (PCOS), and which model could be used in future research? DESIGN PCOS models induced by letrozole, letrozole plus high-fat diet (LE+HFD), testosterone propionate (TP) or testosterone propionate plus HFD (TP+HFD) were established in rats. Body weight, energy intake, blood glucose, sex hormone concentrations, lipid profiles and the oestrus cycle were observed. Histology of ovaries, large intestine and fat was displayed. Protein and mRNA levels relating to hormone synthesis, oocyte maturation, the gut barrier, lipid metabolism and inflammation were evaluated using western blotting, immunohistochemistry and PCR. The composition of the microbial community was measured using 16S RNA sequencing. RESULTS Letrozole treatment induced hyperandrogenaemia, polycystic ovarian morphology, a disrupted oestrus cycle and impaired ovarian function, which could be restored within 42 days. Concurrently, letrozole disturbed glucose, fat, and energy metabolism, affected the inflammatory state and compromised intestinal homeostasis. HFD could amplify the disturbances in the metabolism and intestinal microenvironment, and the pituitary-ovarian axis was more efficiently and consistently affected by testosterone propionate. Testosterone propionate and TP+HFD treatment also disturbed the intestinal microenvironment. Although the metabolic effects of testosterone propionate were not as profound as those of letrozole, they were enhanced by HFD. CONCLUSIONS Letrozole is useful for studies on metabolic disturbances in PCOS, and LE+HFD treatment is suitable for investigations on PCOS metabolic abnormalities and the gut-PCOS link. Testosterone propionate injection is appropriate for studying reproductive abnormalities in PCOS, while TP+HFD treatment is the most comprehensive for studying PCOS reproductive abnormalities, metabolic disturbances and the gut-PCOS link.
Collapse
Affiliation(s)
- Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoxu Dong
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenwen Ma
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kunkun Song
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohu Xu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Wu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingmin Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
7
|
Hernández-Munive AK, Molina-Leonor MB, Ayala-González BD, Vázquez-Andrade J, Medina-Nieto A, Fernández-Guasti A. Diabetes mellitus and female sexual response: what do animal models tell us? Sex Med Rev 2024; 12:542-550. [PMID: 38879347 DOI: 10.1093/sxmrev/qeae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND One of the less explored effects of diabetes mellitus (DM) is female sexual dysfunction. Females of different species have been used as models. AIM To analyze the information of animal models of DM and female sexual response (FSR). METHODS The literature of FSR in models of DM was reviewed. OUTCOMES Paradigm- and diabetes-dependent changes have been found in various aspects of the FSR. RESULTS Females in a type 1 DM (DM1) model show a decrease in the number of proestrus events, and ovariectomized females treated with sex hormones have been used. In these females, a reduction in lordosis has been reported; in proceptivity, the data are contradictory. These females present a decrease in sexual motivation that was restored after exogenous insulin. In the type 2 DM (DM2) model, females show regular estrous cycles, normal levels of lordosis behavior, and, depending on the paradigm, decreased proceptivity. These females display normal preference for sexually active males or their olfactory cues when having free physical contact; they lose this preference when tested in paradigms where physical interaction is precluded. CLINICAL TRANSLATION Preclinical data showing the high deleterious effects of a DM1 model and the less drastic effects under a DM2 model are in accordance with clinical data revealing a much higher prevalence of sexual dysfunction in women with DM1 than DM2. STRENGTHS AND LIMITATIONS The main strength is the analysis of the changes in various components of FSR in 2 models of DM. The main limitation is the difficulty in extrapolating the data on FSR from rats to women and that most studies focus on evaluating the impact of severe or chronic-moderate hyperglycemia/hyperinsulinemia on the sexual response, without considering other pathophysiologic alterations generated by DM. CONCLUSION Females with severe hyperglycemia have a decrease in FSR, while those with moderate hyperglycemia show much less drastic effects.
Collapse
Affiliation(s)
- Abigail Karina Hernández-Munive
- Departamento de Farmacobiología, Centro de investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 14330, México City, México
- División de Neurociencias, Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, México City, México
| | | | - Brenda Denisse Ayala-González
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Joanna Vázquez-Andrade
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Alberto Medina-Nieto
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Alonso Fernández-Guasti
- Departamento de Farmacobiología, Centro de investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 14330, México City, México
| |
Collapse
|
8
|
Yang Q, Wuliu J, Zeng L, Huang J, Tang G, Zhang J, Liao K, Deng K. Association between a body shape index and female infertility: a cross-sectional study. BMC Womens Health 2024; 24:486. [PMID: 39227849 PMCID: PMC11373282 DOI: 10.1186/s12905-024-03335-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND The relationship between A Body Shape Index (ABSI) and female infertility is not well understood. ABSI, a novel anthropometric measure, is gaining recognition for its ability to more accurately capture visceral fat characteristics than traditional metrics like BMI. This study aims to explore the association between ABSI and female infertility, considering its potential applications in medical screening and risk assessment. METHODS This cross-sectional study analyzed data from the NHANES from 2013 to 2020. Female infertility was assessed through reproductive health questionnaires, and ABSI was calculated using waist circumference, BMI, and height. Weighted logistic regression models and trend tests were used to evaluate the association between ABSI and female infertility. Restricted cubic splines (RCS) were employed to explore potential nonlinear relationships. Subgroup analyses were conducted to examine the consistency of the association across various demographic and health-related factors. Sensitivity analyses were also performed, including the exclusion of participants with missing covariate data, the application of propensity score matching, and restricting the analysis to women aged 20-45 years. RESULTS The study included 3,718 participants, 433 of whom were diagnosed with infertility. Higher ABSI was associated with an increased risk of female infertility (OR = 1.56, 95% CI: 1.21-2.00, P = 0.001), as demonstrated by weighted logistic regression and trend tests. Women in the highest ABSI quartile had a significantly higher prevalence of infertility compared to those in the lowest quartile (OR = 1.73, 95% CI: 1.27-2.37, P = 0.001). RCS curves indicated a linear positive relationship between ABSI and infertility risk, with a critical value at 0.079. Subgroup and sensitivity analyses confirmed the stability of these findings. CONCLUSION This study demonstrates a positive linear relationship between ABSI and the risk of female infertility. The use of a simple, non-invasive ABSI measurement could facilitate the early identification of high-risk individuals in large-scale screenings, potentially helping to prevent or reduce the incidence of infertility.
Collapse
Affiliation(s)
- Qian Yang
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Jianxiong Wuliu
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Lingling Zeng
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Jinfa Huang
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Guihua Tang
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Junchao Zhang
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China
| | - Kedan Liao
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China.
| | - Kaixian Deng
- Department of Gynecology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan, 528308, PR China.
| |
Collapse
|
9
|
Norazman SI, Mohd Zaffarin AS, Shuid AN, Hassan H, Soleiman IN, Kuan WS, Alias E. A Review of Animal Models for Studying Bone Health in Type-2 Diabetes Mellitus (T2DM) and Obesity. Int J Mol Sci 2024; 25:9399. [PMID: 39273348 PMCID: PMC11394783 DOI: 10.3390/ijms25179399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Preclinical research on diabetes and obesity has been carried out in various animal models over the years. These animal models are developed from genetic manipulation that affects their body metabolism, chemical-induced procedures, diet alteration/modifications, or combinations of the aforementioned approaches. The diabetic and obesity animal models have allowed researchers to not only study the pathological aspect of the diseases but also enable them to screen and explore potential therapeutic compounds. Besides several widely known complications such as macrovascular diseases, diabetic neuropathy, nephropathy and retinopathy, type 2 diabetes mellitus is also known to affect bone health. There is also evidence to suggest obesity affects bone health. Therefore, continuous research needs to be conducted to find a remedy or solution to this matter. Previous literature reported evidence of bone loss in animal models of diabetes and obesity. These findings, as highlighted in this review, further augment the suggestion of an inter-relationship between diabetes, obesity and bone loss.
Collapse
Affiliation(s)
- Saiful Iqbal Norazman
- The Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Anis Syauqina Mohd Zaffarin
- The Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA, Sg Buloh 47000, Malaysia
| | - Haniza Hassan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Malaysia
| | - Ima Nirwana Soleiman
- The Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Wong Sok Kuan
- The Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| | - Ekram Alias
- The Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
10
|
Abbott DH, Hutcherson BA, Dumesic DA. Anti-Müllerian Hormone: A Molecular Key to Unlocking Polycystic Ovary Syndrome? Semin Reprod Med 2024; 42:41-48. [PMID: 38908381 DOI: 10.1055/s-0044-1787525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Anti-Müllerian hormone (AMH) is an important component within androgen receptor (AR)-regulated pathways governing the hyperandrogenic origin of polycystic ovary syndrome (PCOS). In women with PCOS, granulosa cell AMH overexpression in developing ovarian follicles contributes to elevated circulating AMH levels beginning at birth and continuing in adolescent daughters of PCOS women. A 6 to 7% incidence among PCOS women of gene variants coding for AMH or its receptor, AMHR2, suggests genetic contributions to AMH-related pathogenesis. Discrete gestational AMH administration to pregnant mice induces hypergonadotropic hyperandrogenic, PCOS-like female offspring with high circulating AMH levels that persist over three generations, suggesting epigenetic contributions to PCOS through developmental programming. Moreover, adult-onset, selective hyperactivation of hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH) induces hypergonadotropic hyperandrogenism and PCOS-like traits in female mice. Both gestational and adult AMH inductions of PCOS-like traits are prevented by GnRH antagonist coadministration, implicating luteinizing hormone-dependent ovarian theca cell testosterone (T) action, mediated through the AR in AMH-induced pathogenesis. Interestingly, gestational or peripubertal exogenous T or dihydrotestosterone induction of PCOS-like traits in female mice, rats, sheep, and monkeys fails to elicit ovarian AMH hypersecretion; thus, AMH excess per se may lead to a distinct pathogenic contribution to hyperandrogenic PCOS origins.
Collapse
Affiliation(s)
- David H Abbott
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
| | - Beverly A Hutcherson
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, Wisconsin
- Endocrinology and Reproductive Physiology Training Program, University of Wisconsin, Madison, Wisconsin
- Dean's Office, University of Wisconsin School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Daniel A Dumesic
- Department of Obstetrics and Gynecology, University of California, Los Angeles, California
| |
Collapse
|
11
|
Liu H, Tu M, Yin Z, Zhang D, Ma J, He F. Unraveling the complexity of polycystic ovary syndrome with animal models. J Genet Genomics 2024; 51:144-158. [PMID: 37777062 DOI: 10.1016/j.jgg.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 10/02/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a highly familial and heritable endocrine disorder. Over half of the daughters born to women with PCOS may eventually develop their own PCOS-related symptoms. Progress in the treatment of PCOS is currently hindered by the complexity of its clinical manifestations and incomplete knowledge of its etiopathogenesis. Various animal models, including experimentally induced, naturally occurring, and spontaneously arising ones, have been established to emulate a wide range of phenotypical and pathological traits of human PCOS. These studies have led to a paradigm shift in understanding the genetic, developmental, and evolutionary origins of this disorder. Furthermore, emerging evidence suggests that animal models are useful in evaluating state-of-the-art drugs and treatments for PCOS. This review aims to provide a comprehensive summary of recent studies of PCOS in animal models, highlighting the power of these disease models in understanding the biology of PCOS and aiding high-throughput approaches.
Collapse
Affiliation(s)
- Huanju Liu
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Mixue Tu
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Zhiyong Yin
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Dan Zhang
- Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Zhejiang Provincial Clinical Research Center for Child Health, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Clinical Research Center on Birth Defect Prevention and Intervention of Zhejiang Province, Hangzhou, Zhejiang 310006, China.
| | - Jun Ma
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| | - Feng He
- Center for Genetic Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China; Key Laboratory of Women's Reproductive Health of Zhejiang Province and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorder, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
12
|
Wu H, Zhao B, Yao Q, Kang J. Dehydroepiandrosterone-induced polycystic ovary syndrome mouse model requires continous treatments to maintain reproductive phenotypes. J Ovarian Res 2023; 16:207. [PMID: 37880784 PMCID: PMC10599050 DOI: 10.1186/s13048-023-01299-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/16/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common endocrinopathy associated with infertility and metabolic disorder in women of reproductive age. Animal models have been developed and used as tools to unravel the pathogenesis of PCOS, among which most postnatal models employ continuing experimental manipulations. However, the persistence and stability of these animals after modeling is unknown. Dehydroepiandrosterone (DHEA)-induced PCOS mouse model is commonly used in PCOS studies. Thus the aim of the present study was to investigate the reproductive features of DHEA-induced PCOS mice fed a normal chow or an high-fat diet (HFD) with treatment withdrawal or consecutive treatments after PCOS mouse models were established. METHODS Prepubertal C57BL/6 J mice (age 25 days) were injected (s.c.) daily with DHEA on a normal chow or a 60% HFD for 20 consecutive days to induce PCOS mouse models. Mice injected with the vehicle sesame oil were used as controls. After 20 days, mice were divided into 2 groups, namely "Continue dosing group" and "Stop dosing group". The animals were consecutively treated with DHEA or DHEA + HFD, or housed without any treatment for 2 or 4 weeks. Estrous cycles were evaluated during this period. At the end of the experiment, serum testosterone (T) levels were measured and the morphology of ovaries was evaluated. RESULTS The mice in Continue dosing groups maintained reproductive phenotypes of PCOS mouse models. In contrast, 2 or 4 weeks after PCOS models were established, the mice with treatment withdrawal in Stop dosing groups exhibited normal serum testosterone levels, regular estrous cycle, and relatively normal ovarian morphology. In addition, even with consecutive treatments, there was no marked difference in body weight between DHEA mice on the normal chow or an HFD in Continue dosing groups and the control animals 3 weeks after modeling. CONCLUSIONS After PCOS mice were induced with DHEA or DHEA + HFD, the mice still need consecutive treatments to maintain reproductive phenotypes to be regarded as PCOS mice that meet the diagnostic criteria of PCOS defined by the 2003 Rotterdam criteria.
Collapse
Affiliation(s)
- Haowen Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, No.38 Xueyuan Rd, Haidian District, Beijing, 100191, China
| | - Bining Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, No.38 Xueyuan Rd, Haidian District, Beijing, 100191, China
| | - Qiyang Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, No.38 Xueyuan Rd, Haidian District, Beijing, 100191, China
| | - Jihong Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, No.38 Xueyuan Rd, Haidian District, Beijing, 100191, China.
| |
Collapse
|
13
|
Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14:1015-1037. [PMID: 37401013 PMCID: PMC10445217 DOI: 10.1111/jdi.14034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/05/2023] Open
Abstract
Diabetes mellitus is still expanding globally and is epidemic in developing countries. The combat of this plague has caused enormous economic and social burdens related to a lowered quality of life in people with diabetes. Despite recent significant improvements of life expectancy in patients with diabetes, there is still a need for efforts to elucidate the complexities and mechanisms of the disease processes to overcome this difficult disorder. To this end, the use of appropriate animal models in diabetes studies is invaluable for translation to humans and for the development of effective treatment. In this review, a variety of animal models of diabetes with spontaneous onset in particular will be introduced and discussed for their implication in diabetes research.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Exploratory Medicine for Nature, Life and HumansToho University School of MedicineChibaJapan
- Department of PathologyHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
14
|
Borges JCO, Oliveira VAB, Serdan TDA, Silva FLR, Santos CS, Pauferro JRB, Ribas ASF, Manoel R, Pereira ACG, Correa IS, Pereira JNB, Bazotte RB, Levada-Pires AC, Pithon-Curi TC, Gorjão R, Curi R, Hirabara SM, Masi LN. Brain glucose hypometabolism and hippocampal inflammation in Goto-Kakizaki rats. Braz J Med Biol Res 2023; 56:e12742. [PMID: 37377307 DOI: 10.1590/1414-431x2023e12742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/05/2023] [Indexed: 06/29/2023] Open
Abstract
Brain glucose hypometabolism and neuroinflammation are early pathogenic manifestations in neurological disorders. Neuroinflammation may also disrupt leptin signaling, an adipokine that centrally regulates appetite and energy balance by acting on the hypothalamus and exerting neuroprotection in the hippocampus. The Goto-Kakizaki (GK) rat is a non-obese type 2 diabetes mellitus (T2DM) animal model used to investigate diabetes-associated molecular mechanisms without obesity jeopardizing effects. Wistar and GK rats received the maintenance adult rodent diet. Also, an additional control group of Wistar rats received a high-fat and high-sugar diet (HFHS) provided by free consumption of condensed milk. All diets and water were provided ad libitum for eight weeks. Brain glucose uptake was evaluated by 2-deoxy-2-[fluorine-18] fluoro-D-glucose under basal (saline administration) or stimulated (CL316,243, a selective β3-AR agonist) conditions. The animals were fasted for 10-12 h, anesthetized, and euthanized. The brain was quickly dissected, and the hippocampal area was sectioned and stored at -80°C in different tubes for protein and RNA analyses on the same animal. GK rats exhibited attenuated brain glucose uptake compared to Wistar animals and the HFHS group under basal conditions. Also, the hippocampus of GK rats displayed upregulated leptin receptor, IL-1β, and IL-6 gene expression and IL-1β and the subunit of the transcription factor NF-κB (p-p65) protein expression. No significant alterations were detected in the hippocampus of HFHS rats. Our data indicated that a genetic predisposition to T2DM has significant brain deteriorating features, including brain glucose hypometabolism, neuroinflammation, and leptin signaling disruption in the hippocampal area.
Collapse
Affiliation(s)
- J C O Borges
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - V A B Oliveira
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - T D A Serdan
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - F L R Silva
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - C S Santos
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - J R B Pauferro
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - A S F Ribas
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Manoel
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - A C G Pereira
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - I S Correa
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - J N B Pereira
- Seção de Produção de Imunobiológicos, Bioindustrial Centro, Instituto Butantan, São Paulo, SP, Brasil
| | - R B Bazotte
- Programa de Pós-Graduação em Ciências Farmacêuticas, Departamento de Farmacologia e Terapêutica, Universidade Estadual de Maringá, Maringá, PR, Brasil
| | - A C Levada-Pires
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - T C Pithon-Curi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Gorjão
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - R Curi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
- Seção de Produção de Imunobiológicos, Bioindustrial Centro, Instituto Butantan, São Paulo, SP, Brasil
| | - S M Hirabara
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
| | - L N Masi
- Programa Interdisciplinar de Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, SP, Brasil
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brasil
| |
Collapse
|
15
|
Yamamoto Y, Ura K, Matsukawa T, Saita T, Shin M. Immunohistochemical Localization of Alogliptin, a DPP-4 Inhibitor, in Tissues of Normal and Type 2 Diabetes Model Rat. Acta Histochem Cytochem 2022; 55:185-192. [PMID: 36688140 PMCID: PMC9840470 DOI: 10.1267/ahc.22-00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/23/2022] [Indexed: 12/15/2022] Open
Abstract
We investigated the pharmacokinetics of alogliptin (AG) at the cell and tissue level in healthy Wistar rats and a type 2 diabetic Goto-Kakizaki (GK) rat model. Immunohistochemistry of the renal tissue in these rats, post 1 hr of AG administration, showed that the signal was observed in the glomeruli, proximal tubule S3 segments, distal tubules, collecting ducts, and only in the brush border of the epithelial cells of the proximal tubule S1, S2 segments. After 6 hr of AG administration, the staining intensity of the regions other than the S3 segments was considerably reduced in Wistar rats, with no change observed in GK rats. At 24 hr, the staining intensity was considerably reduced, even in GK rats; however, the staining of the S3 segment remained unaltered in both. Hepatocytes in zone III of the hepatic lobule were more intensely stained than those in zone I in Wistar rats at 1 hr. However, almost no staining was observed in the hepatocytes of GK rats at 1 hr. Complete loss of signal was observed in the hepatocytes of the Wistar rats after 6 hr. This study revealed that the pharmacokinetics of AG in GK rats are different from those in Wistar rats.
Collapse
Affiliation(s)
- Yutaro Yamamoto
- Department of Applied Life Science, Faculty of Biotechnology and Life Science, Sojo University, 4–22–1 Ikeda, Nishi-ku, Kumamoto 860–0082, Japan
| | - Kanae Ura
- Department of Applied Life Science, Faculty of Biotechnology and Life Science, Sojo University, 4–22–1 Ikeda, Nishi-ku, Kumamoto 860–0082, Japan
| | - Takuma Matsukawa
- Department of Applied Life Science, Faculty of Biotechnology and Life Science, Sojo University, 4–22–1 Ikeda, Nishi-ku, Kumamoto 860–0082, Japan
| | - Tetsuya Saita
- Department of Applied Life Science, Faculty of Biotechnology and Life Science, Sojo University, 4–22–1 Ikeda, Nishi-ku, Kumamoto 860–0082, Japan
| | - Masashi Shin
- Department of Applied Life Science, Faculty of Biotechnology and Life Science, Sojo University, 4–22–1 Ikeda, Nishi-ku, Kumamoto 860–0082, Japan
| |
Collapse
|
16
|
Wang R, Gu Z, Wang Y, Yin X, Liu W, Chen W, Huang Y, Wu J, Yang S, Feng L, Zhou L, Li L, Di W, Pu X, Huang L, Qian K. A “One‐Stop Shop” Decision Tree for Diagnosing and Phenotyping Polycystic Ovarian Syndrome on Serum Metabolic Fingerprints. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202206670] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Indexed: 01/06/2025]
Abstract
AbstractPolycystic ovary syndrome (PCOS) is a common endocrine disease regulated by metabolic disorders, the effective intervention of which depends on diverse phenotypes (e.g., insulin resistance). Serum metabolic fingerprint (SMF) holds promise in characterizing the pathogenesis stress related to diseases; yet, PCOS diagnosis and phenotyping are time‐consuming and challenging due to the lack of an integrated metabolic tool. Here, a nanoparticle‐enhanced laser desorption/ionization mass spectrometry platform is introduced for one‐time serum metabolic fingerprinting and to identify the metabolic heterogeneity associated with obesity in PCOS patients. A decision tree based on the acquired SMFs is constructed, and real‐world simulations on independent internal and external cohorts are performed. The decision tree yields the area under the receiver operating characteristic curves (AUC) of 0.967 for PCOS diagnosis and AUC of 0.898 for phenotyping, respectively. The technical robustness of the “one‐stop shop” decision tree across laboratories is validated for clinical utility. The decision tree aims to improve PCOS management in comparison to clinical assessment, leading to a potential reduction in multiple blood tests and physician workload.
Collapse
Affiliation(s)
- Ruimin Wang
- Department of Clinical Laboratory Medicine Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai 200030 P. R. China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai 200030 P. R. China
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Zhuowei Gu
- Shanghai Key Laboratory of Gynecologic Oncology Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai 200127 P. R. China
- Department of Obstetrics and Gynecology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 P. R. China
| | - Yuan Wang
- Center for Reproductive Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai P. R. China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics Shanghai P. R. China
| | - Xia Yin
- Shanghai Key Laboratory of Gynecologic Oncology Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai 200127 P. R. China
- Department of Obstetrics and Gynecology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 P. R. China
| | - Wanshan Liu
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Wei Chen
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Yida Huang
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Jiao Wu
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Shouzhi Yang
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Lei Feng
- Instrumental Analysis Center Shanghai Jiao Tong University No. 800 Dongchuan Road Shanghai 201100 P. R. China
| | - Li Zhou
- Beijing Health Biotech Co. Ltd. No. 7, Science Park Road, Changping District Beijing P. R. China
| | - Lin Li
- Beijing Health Biotech Co. Ltd. No. 7, Science Park Road, Changping District Beijing P. R. China
| | - Wen Di
- Shanghai Key Laboratory of Gynecologic Oncology Renji Hospital School of Medicine Shanghai Jiaotong University Shanghai 200127 P. R. China
- Department of Obstetrics and Gynecology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 P. R. China
| | - Xiaowen Pu
- Shanghai First Maternity and Infant Hospital Tongji University School of Medicine Shanghai 201204 P. R. China
| | - Lin Huang
- Department of Clinical Laboratory Medicine Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai 200030 P. R. China
- Shanghai Institute of Thoracic Oncology Shanghai Chest Hospital Shanghai Jiao Tong University Shanghai 200030 P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes School of Biomedical Engineering Institute of Medical Robotics and Med‐X Research Institute Shanghai Jiao Tong University Shanghai 200030 P. R. China
| |
Collapse
|
17
|
Abstract
Polycystic ovary syndrome (PCOS) is a complex disease affecting up to 15% of women of reproductive age. Women with PCOS suffer from reproductive dysfunctions with excessive androgen secretion and irregular ovulation, leading to reduced fertility and pregnancy complications. The syndrome is associated with a wide range of comorbidities including type 2 diabetes, obesity, and psychiatric disorders. Despite the high prevalence of PCOS, its etiology remains unclear. To understand the pathophysiology of PCOS, how it is inherited, and how to predict PCOS, and prevent and treat women with the syndrome, animal models provide an important approach to answering these fundamental questions. This minireview summarizes recent investigative efforts on PCOS-like rodent models aiming to define underlying mechanisms of the disease and provide guidance in model selection. The focus is on new genetic rodent models, on a naturally occurring rodent model, and provides an update on prenatal and peripubertal exposure models.
Collapse
|
18
|
Anti-Müllerian Hormone and Polycystic Ovary Syndrome in Women and Its Male Equivalent. Biomedicines 2022; 10:biomedicines10102506. [PMID: 36289767 PMCID: PMC9599141 DOI: 10.3390/biomedicines10102506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
This article reviews the main findings on anti-Müllerian hormone (AMH) and its involvement in the pathogenesis of polycystic ovary syndrome (PCOS) and its male equivalent. In women, AMH is produced by granulosa cells from the mid-fetal life to menopause and is a reliable indirect marker of ovarian reserve. AMH protects follicles from atresia, inhibits their differentiation in the ovary, and stimulates gonadotrophin-releasing hormone neurons pulsatility. AMH overexpression in women with PCOS likely contributes to the increase of the follicle cohort and of androgen levels, leading to follicular arrest and anovulation. In the male, AMH is synthesized at high levels by Sertoli cells from fetal life to puberty when serum AMH falls to levels similar to those observed in women. AMH is involved in the differentiation of the genital tract during fetal life and plays a role in Sertoli and Leydig cells differentiation and function. Serum AMH is used to assess Sertoli cell function in children with disorders of sex development and various conditions affecting the hypothalamic–pituitary–testicular axis. Although the reproductive function of male relative of women with PCOS has been poorly investigated, adolescents have elevated levels of AMH which could play a detrimental role on their fertility.
Collapse
|
19
|
Seal SV, Henry M, Pajot C, Holuka C, Bailbé D, Movassat J, Darnaudéry M, Turner JD. A Holistic View of the Goto-Kakizaki Rat Immune System: Decreased Circulating Immune Markers in Non- Obese Type 2 Diabetes. Front Immunol 2022; 13:896179. [PMID: 35677049 PMCID: PMC9168276 DOI: 10.3389/fimmu.2022.896179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Type-2 diabetes is a complex disorder that is now considered to have an immune component, with functional impairments in many immune cell types. Type-2 diabetes is often accompanied by comorbid obesity, which is associated with low grade inflammation. However,the immune status in Type-2 diabetes independent of obesity remains unclear. Goto-Kakizaki rats are a non-obese Type-2 diabetes model. The limited evidence available suggests that Goto-Kakizaki rats have a pro-inflammatory immune profile in pancreatic islets. Here we present a detailed overview of the adult Goto-Kakizaki rat immune system. Three converging lines of evidence: fewer pro-inflammatory cells, lower levels of circulating pro-inflammatory cytokines, and a clear downregulation of pro-inflammatory signalling in liver, muscle and adipose tissues indicate a limited pro-inflammatory baseline immune profile outside the pancreas. As Type-2 diabetes is frequently associated with obesity and adipocyte-released inflammatory mediators, the pro-inflammatory milieu seems not due to Type-2 diabetes per se; although this overall reduction of immune markers suggests marked immune dysfunction in Goto-Kakizaki rats.
Collapse
Affiliation(s)
- Snehaa V Seal
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Mathilde Henry
- Institut National de Recherche Pour l'agriculture, l'alimentation et l'environnement (INRAE), Bordeaux Institut National Polytechnique (INP), NutriNeuro, Unité Mixte de Recherche (UMR) 1286, University of Bordeaux, Bordeaux, France
| | - Clémentine Pajot
- Institut National de Recherche Pour l'agriculture, l'alimentation et l'environnement (INRAE), Bordeaux Institut National Polytechnique (INP), NutriNeuro, Unité Mixte de Recherche (UMR) 1286, University of Bordeaux, Bordeaux, France
| | - Cyrielle Holuka
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Danielle Bailbé
- Université de Paris, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Centre National de la Recherche Scientifique -Unité Mixte de Recherche (CNRS UMR) 8251, Paris, France
| | - Jamileh Movassat
- Université de Paris, Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptative), Centre National de la Recherche Scientifique -Unité Mixte de Recherche (CNRS UMR) 8251, Paris, France
| | - Muriel Darnaudéry
- Institut National de Recherche Pour l'agriculture, l'alimentation et l'environnement (INRAE), Bordeaux Institut National Polytechnique (INP), NutriNeuro, Unité Mixte de Recherche (UMR) 1286, University of Bordeaux, Bordeaux, France
| | - Jonathan D Turner
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| |
Collapse
|
20
|
Heather LC, Hafstad AD, Halade GV, Harmancey R, Mellor KM, Mishra PK, Mulvihill EE, Nabben M, Nakamura M, Rider OJ, Ruiz M, Wende AR, Ussher JR. Guidelines on Models of Diabetic Heart Disease. Am J Physiol Heart Circ Physiol 2022; 323:H176-H200. [PMID: 35657616 PMCID: PMC9273269 DOI: 10.1152/ajpheart.00058.2022] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Diabetes is a major risk factor for cardiovascular diseases, including diabetic cardiomyopathy, atherosclerosis, myocardial infarction, and heart failure. As cardiovascular disease represents the number one cause of death in people with diabetes, there has been a major emphasis on understanding the mechanisms by which diabetes promotes cardiovascular disease, and how antidiabetic therapies impact diabetic heart disease. With a wide array of models to study diabetes (both type 1 and type 2), the field has made major progress in answering these questions. However, each model has its own inherent limitations. Therefore, the purpose of this guidelines document is to provide the field with information on which aspects of cardiovascular disease in the human diabetic population are most accurately reproduced by the available models. This review aims to emphasize the advantages and disadvantages of each model, and to highlight the practical challenges and technical considerations involved. We will review the preclinical animal models of diabetes (based on their method of induction), appraise models of diabetes-related atherosclerosis and heart failure, and discuss in vitro models of diabetic heart disease. These guidelines will allow researchers to select the appropriate model of diabetic heart disease, depending on the specific research question being addressed.
Collapse
Affiliation(s)
- Lisa C Heather
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anne D Hafstad
- Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ganesh V Halade
- Department of Medicine, The University of Alabama at Birmingham, Tampa, Florida, United States
| | - Romain Harmancey
- Department of Internal Medicine, Division of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Miranda Nabben
- Departments of Genetics and Cell Biology, and Clinical Genetics, Maastricht University Medical Center, CARIM School of Cardiovascular Diseases, Maastricht, the Netherlands
| | - Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Oliver J Rider
- University of Oxford Centre for Clinical Magnetic Resonance Research, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Matthieu Ruiz
- Montreal Heart Institute, Montreal, Quebec, Canada.,Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Adam R Wende
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.,Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
21
|
Li MX, Li MY, Lei JX, Wu YZ, Li ZH, Chen LM, Zhou CL, Su JY, Huang GX, Huang XQ, Zheng XB. Huangqin decoction ameliorates DSS-induced ulcerative colitis: Role of gut microbiota and amino acid metabolism, mTOR pathway and intestinal epithelial barrier. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154052. [PMID: 35344714 DOI: 10.1016/j.phymed.2022.154052] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND The clinical treatment of ulcerative colitis (UC) is limited. A traditional Chinese medicinal formula, Huangqin decoction (HQD), is chronicled in Shang Han Lun and is widely used to ameliorate gastrointestinal disorders, such as UC; however, its mechanism is yet to be clarified. PURPOSE The present study aimed to investigate the effect of HQD on 7-day colitis induced by 3% dextran sulfate sodium (DSS) in mice and further explore the inhibitory effect of metabolites on DSS-damaged FHC cells. METHODS The therapeutic efficacy of HQD was evaluated in a well-established DSS-induced colitis mice model. The clinical symptoms were analyzed, and biological samples were collected for microscopic examination, metabolomics, metagenomics, and the evaluation of the epithelial barrier function. The mechanism of metabolites regulated by HQD was evaluated in the DSS-induced FHC cell damage model. The samples were collected to detect the physiological functions of the cells. RESULTS HQD suppressed the inflammation of DSS-induced colitis in vivo, attenuated DSS-induced clinical manifestations, reversed colon length reduction, and reduced histological injury. After HQD treatment, the DSS-induced gut dysbiosis was modulated, and the gut microbiota achieved a new equilibrium state. In addition, HQD activated the mTOR signaling pathway by upregulating amino acid metabolism. Significant phosphorylation of S6 and 4E-BP1 ameliorated intestinal epithelial barrier dysfunction. Moreover, HQD-regulated metabolites protected the epithelial barrier integrity by inhibiting DSS-induced apoptosis of FHC cells and regulating the proteins affecting apoptosis and cell-cell junction. CONCLUSIONS These findings indicated that the mechanism of HQD was related to regulating the gut microbiota and amino acid metabolism, activating the mTOR signaling pathway, and protecting the intestinal mucosal barrier integrity.
Collapse
Affiliation(s)
- Mu-Xia Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Min-Yao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Jun-Xuan Lei
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Yu-Zhu Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Ze-Hao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Lin-Ming Chen
- Guangzhou Huibiao Testing Technology Center, Guangzhou 510700, P.R. China
| | | | - Ji-Yan Su
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan 528000, Guangdong, P.R. China
| | - Guo-Xin Huang
- Clinical research center, Shantou central hospital, Shantou 515041, China
| | - Xiao-Qi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.
| | - Xue-Bao Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.
| |
Collapse
|
22
|
Role of insulin resistance on fertility–focus on polycystic ovary syndrome. ANNALES D'ENDOCRINOLOGIE 2022; 83:199-202. [DOI: 10.1016/j.ando.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
23
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
24
|
Benammar A, Derisoud E, Vialard F, Palmer E, Ayoubi JM, Poulain M, Chavatte-Palmer P. The Mare: A Pertinent Model for Human Assisted Reproductive Technologies? Animals (Basel) 2021; 11:2304. [PMID: 34438761 PMCID: PMC8388489 DOI: 10.3390/ani11082304] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/28/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022] Open
Abstract
Although there are large differences between horses and humans for reproductive anatomy, follicular dynamics, mono-ovulation, and embryo development kinetics until the blastocyst stage are similar. In contrast to humans, however, horses are seasonal animals and do not have a menstrual cycle. Moreover, horse implantation takes place 30 days later than in humans. In terms of artificial reproduction techniques (ART), oocytes are generally matured in vitro in horses because ovarian stimulation remains inefficient. This allows the collection of oocytes without hormonal treatments. In humans, in vivo matured oocytes are collected after ovarian stimulation. Subsequently, only intra-cytoplasmic sperm injection (ICSI) is performed in horses to produce embryos, whereas both in vitro fertilization and ICSI are applied in humans. Embryos are transferred only as blastocysts in horses. In contrast, four cells to blastocyst stage embryos are transferred in humans. Embryo and oocyte cryopreservation has been mastered in humans, but not completely in horses. Finally, both species share infertility concerns due to ageing and obesity. Thus, reciprocal knowledge could be gained through the comparative study of ART and infertility treatments both in woman and mare, even though the horse could not be used as a single model for human ART.
Collapse
Affiliation(s)
- Achraf Benammar
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Emilie Derisoud
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - François Vialard
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| | - Eric Palmer
- Académie d’Agriculture de France, 75007 Paris, France;
| | - Jean Marc Ayoubi
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Marine Poulain
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
- Department of Gynaecology and Obstetrics, Foch Hospital, 92150 Suresnes, France
| | - Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, 78350 Jouy-en-Josas, France; (A.B.); (E.D.); (F.V.); (J.M.A.); (M.P.)
- Ecole Nationale Vétérinaire d’Alfort, BREED, 94700 Maisons-Alfort, France
| |
Collapse
|