1
|
Silva BTDA, Martins-Perles JVC, Bossolani GDP, Lima MM, Sehaber-Sierakowski CC, Gremaschi LB, Cunha JPSE, Bersani-Amado CA, Zanoni JN. Quercetin and ibuprofen combination displayed anti-inflammatory effects and also extenuates the enteric neurons damage of arthritic rats. AN ACAD BRAS CIENC 2024; 96:e20230244. [PMID: 39140520 DOI: 10.1590/0001-3765202420230244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/09/2024] [Indexed: 08/15/2024] Open
Abstract
This study aimed to investigate the antioxidant and anti-inflammatory properties of quercetin on the cellular components of the Enteric Nervous System in the ileum of rats with arthritis. Rats were distributed into five groups: control (C), arthritic (AIA), arthritic treated with ibuprofen (AI), arthritic treated with quercetin (AQ) and arthritic treated with both ibuprofen and quercetin (AIQ). The ileum was processed for immunohistochemical techniques for HuC/D, calcitonin gene-related peptide, and vasoactive intestinal polypeptide. Measurements in histological sections, chemiluminescence assays, and total antioxidant capacity were also performed. Rheumatoid arthritis resulted in a decrease in neuronal density, yet neuroplasticity mechanisms were evident through observed changes in varicosities size and neuronal area compared to the control group. Reduced paw edema and neuroprotective effects were predominantly noted in both plexuses, as evidenced by the increased density preservation of HuC/D-IR neurons in the AIQ group. The increase of lipoperoxidation levels and paw edema volume in the AQ group was observed compared to the arthritic, whereas the AIQ group mainly showed similar results to those observed in the control. The enteropathy associated with arthritis proved to be significant in the field of gastroenterology, and the combination of quercetin and ibuprofen demonstrated promising anti-inflammatory and neuroprotective effects.
Collapse
Affiliation(s)
- Bruna Thais DA Silva
- Universidade Estadual de Maringá, Departamento de Biologia, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | | | - Gleison Daion P Bossolani
- Universidade Estadual de Maringá, Departamento de Farmácia, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - Mariana M Lima
- Universidade Estadual de Maringá, Departamento de Farmácia, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - Camila C Sehaber-Sierakowski
- Universidade Estadual de Maringá, Departamento de Farmácia, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - Lucas B Gremaschi
- Universidade Estadual de Maringá, Departamento de Medicina, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - João Paulo Silveira E Cunha
- Universidade Estadual de Maringá, Departamento de Ciências, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - Ciomar A Bersani-Amado
- Universidade Estadual de Maringá, Departamento de Farmacologia e Terapêutica, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| | - Jacqueline N Zanoni
- Universidade Estadual de Maringá, Departamento de Ciências, Avenida Colombo, 5790, 87020-900 Maringá, PR, Brazil
| |
Collapse
|
2
|
Llorián-Salvador M, Cabeza-Fernández S, Gomez-Sanchez JA, de la Fuente AG. Glial cell alterations in diabetes-induced neurodegeneration. Cell Mol Life Sci 2024; 81:47. [PMID: 38236305 PMCID: PMC10796438 DOI: 10.1007/s00018-023-05024-y] [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: 08/20/2023] [Revised: 10/09/2023] [Accepted: 10/29/2023] [Indexed: 01/19/2024]
Abstract
Type 2 diabetes mellitus is a global epidemic that due to its increasing prevalence worldwide will likely become the most common debilitating health condition. Even if diabetes is primarily a metabolic disorder, it is now well established that key aspects of the pathogenesis of diabetes are associated with nervous system alterations, including deleterious chronic inflammation of neural tissues, referred here as neuroinflammation, along with different detrimental glial cell responses to stress conditions and neurodegenerative features. Moreover, diabetes resembles accelerated aging, further increasing the risk of developing age-linked neurodegenerative disorders. As such, the most common and disabling diabetic comorbidities, namely diabetic retinopathy, peripheral neuropathy, and cognitive decline, are intimately associated with neurodegeneration. As described in aging and other neurological disorders, glial cell alterations such as microglial, astrocyte, and Müller cell increased reactivity and dysfunctionality, myelin loss and Schwann cell alterations have been broadly described in diabetes in both human and animal models, where they are key contributors to chronic noxious inflammation of neural tissues within the PNS and CNS. In this review, we aim to describe in-depth the common and unique aspects underlying glial cell changes observed across the three main diabetic complications, with the goal of uncovering shared glial cells alterations and common pathological mechanisms that will enable the discovery of potential targets to limit neuroinflammation and prevent neurodegeneration in all three diabetic complications. Diabetes and its complications are already a public health concern due to its rapidly increasing incidence, and thus its health and economic impact. Hence, understanding the key role that glial cells play in the pathogenesis underlying peripheral neuropathy, retinopathy, and cognitive decline in diabetes will provide us with novel therapeutic approaches to tackle diabetic-associated neurodegeneration.
Collapse
Affiliation(s)
- María Llorián-Salvador
- Diabetes and Metabolism Research Unit, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University, Belfast, UK.
| | - Sonia Cabeza-Fernández
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain
| | - Jose A Gomez-Sanchez
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain
| | - Alerie G de la Fuente
- Institute for Health and Biomedical Research of Alicante (ISABIAL), Alicante, Spain.
- Institute of Neuroscience CSIC-UMH, San Juan de Alicante, Spain.
| |
Collapse
|
3
|
Bagyánszki M, Bódi N. Key elements determining the intestinal region-specific environment of enteric neurons in type 1 diabetes. World J Gastroenterol 2023; 29:2704-2716. [PMID: 37274063 PMCID: PMC10237112 DOI: 10.3748/wjg.v29.i18.2704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Diabetes, as a metabolic disorder, is accompanied with several gastrointestinal (GI) symptoms, like abdominal pain, gastroparesis, diarrhoea or constipation. Serious and complex enteric nervous system damage is confirmed in the background of these diabetic motility complaints. The anatomical length of the GI tract, as well as genetic, developmental, structural and functional differences between its segments contribute to the distinct, intestinal region-specific effects of hyperglycemia. These observations support and highlight the importance of a regional approach in diabetes-related enteric neuropathy. Intestinal large and microvessels are essential for the blood supply of enteric ganglia. Bidirectional morpho-functional linkage exists between enteric neurons and enteroglia, however, there is also a reciprocal communication between enteric neurons and immune cells on which intestinal microbial composition has crucial influence. From this point of view, it is more appropriate to say that enteric neurons partake in multidirectional communication and interact with these key players of the intestinal wall. These interplays may differ from segment to segment, thus, the microenvironment of enteric neurons could be considered strictly regional. The goal of this review is to summarize the main tissue components and molecular factors, such as enteric glia cells, interstitial cells of Cajal, gut vasculature, intestinal epithelium, gut microbiota, immune cells, enteroendocrine cells, pro-oxidants, antioxidant molecules and extracellular matrix, which create and determine a gut region-dependent neuronal environment in diabetes.
Collapse
Affiliation(s)
- Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
4
|
Hao X, Ding N, Zhang Y, Yang Y, Zhao Y, Zhao J, Li Y, Li Z. Benign regulation of the gut microbiota: The possible mechanism through which the beneficial effects of manual acupuncture on cognitive ability and intestinal mucosal barrier function occur in APP/PS1 mice. Front Neurosci 2022; 16:960026. [PMID: 35992924 PMCID: PMC9382294 DOI: 10.3389/fnins.2022.960026] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/11/2022] [Indexed: 11/22/2022] Open
Abstract
Background Gut microbiota dysbiosis and intestinal barrier injury play vital roles in Alzheimer's disease (AD) onset and development. Our previous studies have demonstrated that manual acupuncture (MA) could improve the cognitive abilities of APP/PS1 mice. However, the effect of MA on the intestinal mucosal barrier and the gut microbiota mechanism through which this effect occurs remain to be clarified. Methods In the APP/PS1 manual acupuncture (Am) group, MA was applied in Baihui (GV20), Yintang (GV29), and Zusanli (ST36). Mice in the APP/PS1 antibiotic + manual acupuncture (Aa) group were treated with an antibiotic mixture and MA at the same time. Probiotics were delivered to the APP/PS1 probiotics (Ap) group. Alterations in spatial learning and memory, the gut microbiota, the intestinal barrier function, and the expression of glial fibrillary acidic protein (GFAP), lipopolysaccharide (LPS), and TNF-α were evaluated in each group. Results Compared with the C57BL/6 control (Cc) group, cognitive ability was significantly decreased, the gut microbiota structure was obviously disrupted, intestinal barrier integrity was drastically impaired, and the intestinal inflammatory response was enhanced in the APP/PS1 control (Ac) group (P < 0.01). These changes were reversed by MA and probiotics (P < 0.01 or P < 0.05), whereas antibiotics inhibited the benign regulation by MA (P < 0.01 or P < 0.05). Conclusion Manual acupuncture can benignly modulate gut microbiota dysbiosis, significantly reduce intestinal inflammation, and effectively alleviate the destruction of the intestinal mucosal barrier in APP/PS1 mice, and the effects are comparable to those of probiotics. The gut microbiota may play an important role in the improvement of the cognitive function and intestinal barrier function by MA.
Collapse
Affiliation(s)
- Xin Hao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ning Ding
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yichen Yang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yali Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yiran Li
- International School, Beijing University of Chinese Medicine, Beijing, China
| | - Zhigang Li
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Liu Y, Huang H, Sun Y, Li Y, Luo B, Cui J, Zhu M, Bi F, Chen K, Liu Y. Monosodium Glutamate-Induced Mouse Model With Unique Diabetic Retinal Neuropathy Features and Artificial Intelligence Techniques for Quantitative Evaluation. Front Immunol 2022; 13:862702. [PMID: 35572527 PMCID: PMC9092070 DOI: 10.3389/fimmu.2022.862702] [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: 01/26/2022] [Accepted: 03/17/2022] [Indexed: 11/26/2022] Open
Abstract
Objective To establish an artificial intelligence-based method to quantitatively evaluate subtle pathological changes in retinal nerve cells and synapses in monosodium glutamate (MSG) mice and provide an effective animal model and technique for quantitative evaluation of retinal neurocytopathies. Methods ICR mice were subcutaneously injected with MSG to establish a model of metabolic syndrome. We then established a mouse model of type 1 diabetes, type 2 diabetes, and KKAy mouse model as control. The HE sections of the retina were visualized using an optical microscope. AI technology was used for quantitative evaluation of the retinal lesions in each group of rats. The surface area custom parameters of the retinal nerve fiber layer (RNFL), inner plexiform layer (IPL), inner nuclear layer (INL), and outer plexiform layer (OPL) were defined as SR, SIPL, SINL, and SOPL, respectively. Their heights were defined as HR, HIPL, HINL, and HOPL, and the number of ganglion cells was defined as A. Then, the attention-augmented fully convolutional Unet network was used to segment the retinal HE images, and AI technology to identify retinal neurocytopathies quantitatively. Results The attention-augmented fully convolutional Unet network increased PA and IOU parameters for INL, OPL, RNFL, and ganglion cells and was superior in recognizing fine structures. A quantitative AI identification of the height of each layer of the retina showed that the heights of the IPL and INL of the MSG model were significantly less than those of the control groups; the retinas of the other diabetic models did not exhibit this pathological feature. The RNFLs of type 2 diabetes were thinner, and the characteristics of retinopathy were not obvious in the other animal models. The pathological changes seen on HE images were consistent with the results of the quantitative AI evaluation. Immunohistochemistry results showed that NMDAR2A, GluR2, and NRG1 were significantly downregulated in the retina of MSG mice. Conclusions The MSG retinopathy model is closely associated with neurotransmitter abnormalities and exhibits important characteristics of retinal neurodegeneration, making it suitable for studying retinal neurocytopathies. The AI recognition technology for retinal images established in the present study can be used for the quantitative and objective evaluation of drug efficacy.
Collapse
Affiliation(s)
- Yanfei Liu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Huang
- Beijing Duan-Dian Pharmaceutical Research & Development Co., Ltd., Beijing, China
| | - Yu Sun
- North China University of Technology, Beijing, China
| | - Yiwen Li
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Binyu Luo
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Cui
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Mengmeng Zhu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fukun Bi
- North China University of Technology, Beijing, China
| | - Keji Chen
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- National Clinical Research Centre for Chinese Medicine Cardiology, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
6
|
CD44 fucosylation on bone marrow-derived mesenchymal stem cells enhances homing and promotes enteric nervous system remodeling in diabetic mice. Cell Biosci 2021; 11:118. [PMID: 34193268 PMCID: PMC8243650 DOI: 10.1186/s13578-021-00632-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/18/2021] [Indexed: 01/15/2023] Open
Abstract
Background Diabetes can cause extensive enteric nervous system (ENS) injuries and gastrointestinal motility disorder. In developing possible treatments, researchers have engaged in tissue regeneration engineering with the very promising bone marrow-derived mesenchymal stem cells (BMSCs). However, BMSCs have poor homing ability to the targeted tissues after intravenous injection. Thus, we aimed to investigate whether enhancing the expression of E-selectin ligand on BMSCs could improve their homing ability and subsequently influence their role in ENS remodeling in diabetic mice. Methods First, we constructed the fucosylation modification of CD44 on BMSCs through a fucosyltransferase VII (FTVII) system to generate a Hematopoietic Cell E-/L-selectin Ligand (HCELL) property, a fucosylated sialyllactosaminyl glycovariant of CD44 that potently binds E-selectin. Next, FTVII-modified and unmodified BMSCs labeled with green fluorescent protein (GFP) were injected into diabetic mice through the tail vein to compare their homing ability to the gastrointestinal tract and their effect on ENS remodeling, respectively. A bioluminescent imaging system was used to evaluate the homing ability of GFP-labeled BMSCs with and without FTVII modification, to the gastrointestinal tract. Gastrointestinal motility was assessed by gastrointestinal transient time, defecation frequency, stool water content and colon strips contractility. Immunofluorescence staining and western blotting were used to assess the expression levels of protein gene product 9.5 (PGP9.5), glial fibrillary acidic protein (GFAP) and glial cell line-derived neurotrophic factor (GDNF). Results The FTVII-mediated α(1,3)-fucosylation modification of CD44 on BMSCs generated a HCELL property. Bioluminescent imaging assays showed that FTVII-modified BMSCs had enhanced homing ability to gastrointestinal tract, mainly to the colon, 24 h after injection through the tail vein. Compared with diabetic mice, FTVII-modified BMSCs significantly promoted the gastrointestinal motility and the ENS remodeling, including intestinal peristalsis (P < 0.05), increased feces excretion (P < 0.05) and the water content of the feces (P < 0.05), restored the spontaneous contraction of the colon (P < 0.05), and upregulated the protein expression levels of PGP9.5 (P < 0.01), GFAP (P < 0.001), and GDNF (P < 0.05), while unmodified BMSCs did not (P > 0.05). Conclusions CD44 fucosylation modification on murine BMSCs promotes homing ability to the gastrointestinal tract and ENS remodeling in diabetic mice. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00632-2.
Collapse
|
7
|
Yang X, Wu S, Feng Z, Yi G, Zheng Y, Xia Z. Combination therapy with semaglutide and rosiglitazone as a synergistic treatment for diabetic retinopathy in rodent animals. Life Sci 2021; 269:119013. [PMID: 33417950 DOI: 10.1016/j.lfs.2020.119013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE To investigate the protective efficacies and potent mechanisms of combination therapy with semaglutide and rosiglitazone (RSG) on the high-glucose incubated human ARPE-19 cells and diabetic retinopathy (DR) model rats. MAIN METHODS The CCK-8 methods were used to evaluate the protective effects of semaglutide and RSG alone or combination on the cell viability of high-glucose treated ARPE-19 cells. After the DR rat model was established, the effects of combined treatment on general indexes, retinal morphological changes, retinal Müller cells as well as PI3K/Akt/MTOR related factors of DR model rats were investigated. RESULTS The CCK-8 assay showed obviously enhanced protective efficacies of combination therapy with semaglutide and RSG on the ARPE-19 with oxidative stress induced by high-glucose with combination index all below 1.5 demonstrating obvious synergistic effects. Combined incubation could also effectively decrease the expression of inflammatory factors, including TNF-α, IL-1β, IL-6, and the increase of ROS content in ARPE cell culture supernatant induced by high-glucose. Combined use of the antioxidant, PI3K/Akt and mTOR inhibitors, we further demonstrated that combined incubation of semaglutide and RSG could effectively by reduce high glucose-induced inflammatory injury inhibiting ROS/PI3K/Akt/mTOR signaling. Furthermore, chronic combination treatment effectively improved the histopathological characteristics and down-regulated the GFAP expression in Müller cells as well as PI3K/Akt/MTOR signaling pathway-related factors in retina which was better than any monomer treatment group. CONCLUSIONS Combined semaglutide with RSG exhibited synergistically protective efficacies on retinal cells by decreasing the GFAP expression, inhibiting oxidative stress and PI3K/Akt/MTOR signaling-transduction in DR model rats.
Collapse
Affiliation(s)
- Xiaoxi Yang
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Shuduan Wu
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Zhizhen Feng
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Yuxing Zheng
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China
| | - Zhaoxia Xia
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 51000, PR China.
| |
Collapse
|
8
|
Amer ME, Othamn AI, El-Missiry MA. Melatonin ameliorates diabetes-induced brain injury in rats. Acta Histochem 2021; 123:151677. [PMID: 33401187 DOI: 10.1016/j.acthis.2020.151677] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/30/2023]
Abstract
Diabetic brain is a serious complication of diabetes, and it is associated with oxidative stress and neuronal injury. This study investigated the protective effect of melatonin (MLT) on diabetes-induced brain injury. A rat model of type 2 diabetes mellitus was produced by intraperitoneal injection of nicotinamide 100 mg/kg, followed by intraperitoneal injection of streptozotocin 55 mg/kg. The diabetic rats were orally administered MLT 10 mg/kg of body weight for 15 days. MLT remarkably downregulated serum glucose levels. It also improved levels of the lipid peroxidation product 4-hydroxynonenal, improved levels of antioxidants including glutathione, glutathione peroxidase and glutathione reductase in the brains of the diabetic rats, and this is indicative of the antioxidant potential of MLT. MLT also prevented increase in homocysteine, amyloid-β42 and tau levels in diabetic rats, and this suggests that it can reduce the risk of dementia. This is associated with reduction in the levels of the dopamine, serotonin, and glutamate and is indicative of the regulatory effect of MLT on neurotransmitters. Treatment with MLT improved diabetes-induced structural alteration in the hippocampus and cerebral cortex. MLT significantly reduced caspase-3 and Bax as well as significantly increase Bcl-2 protein and GFAP-positive astrocytes indicating its anti-apoptotic effect. MLT showed remarkable ameliorative effect against biochemical and molecular alterations in the brains of diabetic rats most likely through its antioxidant property.
Collapse
Affiliation(s)
- Maggie E Amer
- Faculty of Science, Mansoura University, Mansoura, Egypt.
| | - Azza I Othamn
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | | |
Collapse
|
9
|
Piovezana Bossolani GD, Silva BT, Colombo Martins Perles JV, Lima MM, Vieira Frez FC, Garcia de Souza SR, Sehaber-Sierakowski CC, Bersani-Amado CA, Zanoni JN. Rheumatoid arthritis induces enteric neurodegeneration and jejunal inflammation, and quercetin promotes neuroprotective and anti-inflammatory actions. Life Sci 2019; 238:116956. [DOI: 10.1016/j.lfs.2019.116956] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
|
10
|
The novel insight into anti-inflammatory and anxiolytic effects of psychobiotics in diabetic rats: possible link between gut microbiota and brain regions. Eur J Nutr 2019; 58:3361-3375. [PMID: 30826905 DOI: 10.1007/s00394-019-01924-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/05/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE Type 2 diabetes mellitus (T2DM) was associated with gut microbial impairment (dysbiosis) and neurological and behavioral disorders. The role of the gut-brain axis in the management of many diseases including T2DM has been the focus of much research activity in the recent years. However, a wide knowledge gap exists about the gut microbial effects on the function of glia cells. Hence, the present study was aimed to examine the effects of psychobatics on dysbiosis and glia cells function in enteric and central nervous system with an inflammatory insight in T2DM. METHODS Thirty rats were treated by Lactobacillus (L.) plantarum, inulin, or their combination (synbiotic) for 8 weeks after inducing T2DM. Fecal sample was collected to evaluate gut microbial composition. Then, the rats were sacrificed, and the colon, amygdala, and prefrontal cortex (PFC) were studied. RESULTS T2DM resulted in dysbiosis and increased levels of glial cell-derived neurotrophic factor (GDNF), glial fibrillary acidic protein (GFAP), and inflammatory markers (IL-17, IL-6, and TLR-2) in the colon and brain. However, concurrent supplementation of L. plantarum and inulin could improve the gut microbial composition as well as reduce the levels of inflammatory cytokines. While the administration of L. plantarum led to a significant decrease in TLR-2 as well as GDNF and GFAP only in the amygdala, the synbiotic intake could make such changes in the colon, amygdala, and PFC. CONCLUSIONS Our findings demonstrated an innovative approach to the beneficial effects of psychobiotics in neuroinflammation and behavioral performance through gut microbiota changes, focusing on possible role of glial cells in gut-brain axis.
Collapse
|
11
|
Trevizan AR, Schneider LCL, Araújo EJDA, Garcia JL, Buttow NC, Nogueira-Melo GDA, Sant'Ana DDMG. Acute Toxoplasma gondii infection alters the number of neurons and the proportion of enteric glial cells in the duodenum in Wistar rats. Neurogastroenterol Motil 2019; 31:e13523. [PMID: 30537037 DOI: 10.1111/nmo.13523] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/16/2018] [Accepted: 11/07/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Toxoplasma gondii infection can occur through the ingestion of raw meat that contains tissue cysts or food that contains oocysts. Through the ingestion of oocysts, the parasite crosses the intestinal barrier, where the enteric nervous system is located. The objective was to investigate the kinetics of neuronal and glial responses during acute T. gondii infection. METHODS We used 45 Wistar rats that were divided into a control group and infected groups that were evaluated at 6, 12, 24, 48, 72 hours, 7 days, 10 days, and 15 days after infection. The rats received 5000 sporulated oocysts of the parasite orally. To detect neurons and enteric glia cells, the myenteric and submucosal plexuses of the duodenum underwent double-labeling immunohistochemical techniques to evaluate HuC/HuD and S100, HuC/HuD and ChAT, and HuC/HuD and nNOS. KEY RESULTS We observed a reduction of the total neuron population in the submucosal plexus 72 hours after infection. Cholinergic neurons decreased in the submucosal plexus 15 days after infection, and nitrergic neurons decreased in the myenteric plexus 72 hours after infection. A decrease in the number of glial cells was observed 7 days after infection in the submucosal plexus, and an increase in the enteric glial cell (EGC)/neuron ratio was found in both plexuses 48 hours after infection. CONCLUSIONS AND INFERENCES We found decrease of neurons and increase in the EGC/neuron ratio in both plexuses caused by acute T. gondii infection, with major alterations 72 hours after oral infection. The number of cholinergic neurons decreased in the submucosal plexus, and the number of nitrergic neurons decreased in the myenteric plexus. A decrease in the number of enteric glial cells was observed in the submucosal plexus, and an increase in the enteric glial cell/neuron ratio was observed in both ganglionate plexuses of the duodenum.
Collapse
|
12
|
Morales-Soto W, Gulbransen BD. Enteric Glia: A New Player in Abdominal Pain. Cell Mol Gastroenterol Hepatol 2018; 7:433-445. [PMID: 30739868 PMCID: PMC6369218 DOI: 10.1016/j.jcmgh.2018.11.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/16/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Chronic abdominal pain is the most common gastrointestinal issue and contributes to the pathophysiology of functional bowel disorders and inflammatory bowel disease. Current theories suggest that neuronal plasticity and broad alterations along the brain-gut axis contribute to the development of chronic abdominal pain, but the specific mechanisms involved in chronic abdominal pain remain incompletely understood. Accumulating evidence implicates glial cells in the development and maintenance of chronic pain. Astrocytes and microglia in the central nervous system and satellite glia in dorsal root ganglia contribute to chronic pain states through reactive gliosis, the modification of glial networks, and the synthesis and release of neuromodulators. In addition, new data suggest that enteric glia, a unique type of peripheral glia found within the enteric nervous system, have the potential to modify visceral perception through interactions with neurons and immune cells. Understanding these emerging roles of enteric glia is important to fully understand the mechanisms that drive chronic pain and to identify novel therapeutic targets. In this review, we discuss enteric glial cell signaling mechanisms that have the potential to influence chronic abdominal pain.
Collapse
Affiliation(s)
| | - Brian D. Gulbransen
- Correspondence Address correspondence to: Brian D. Gulbransen, PhD, Neuroscience Program and Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, Michigan 48824. fax: (517) 355-5125.
| |
Collapse
|
13
|
Luo P, Liu D, Li C, He WX, Zhang CL, Chang MJ. Enteric glial cell activation protects enteric neurons from damage due to diabetes in part via the promotion of neurotrophic factor release. Neurogastroenterol Motil 2018; 30:e13368. [PMID: 29740907 DOI: 10.1111/nmo.13368] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 04/03/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Diabetes can result in pathological changes to enteric nervous system. Our aim was to test the dynamic changes of enteric neurons and identify the role of enteric glial cells (EGCs) in regulating enteric neuron expression in diabetic rats. METHODS A single injection of streptozotocin (STZ) was used to establish diabetic rats. Animals were randomly distributed into diabetic 1-, 4-, 8-, and 16-week groups, as well as age-matched control groups. The PGP9.5- and glial fibrillary acidic protein (GFAP)-immunopositive cells were quantified by immunohistochemistry. The protein levels of PGP9.5, ChAT, nNOS, S-100β, and c-fos were determined by western blotting. The levels of nerve growth factor (NGF), neurotrophin 3 (NT-3), and glial cell-derived neurotrophic factor (GDNF) were tested by ELISA. KEY RESULTS An increase in blood glucose and a decrease in body weight were observed following STZ administration. PGP9.5 expression did not change in the diabetic ileum. However, ChAT increased after 16 weeks, and nNOS decreased after 8 and 16 weeks in the ilea of diabetic rats. The absence of degeneration of enteric neurons during the acute stage of the disease could be the consequence of the up-regulation of GFAP, S-100β, and c-fos. Moreover, the content of NGF, NT-3, and GDNF in the ileum increased by varying degrees after 1 and/or 4 weeks of diabetes. Using 2 co-culture models of EGCs and SH-SY5Y cells in a high glucose condition, the supportive role of EGCs was further confirmed. CONCLUSIONS & INFERENCES Enteric glial cell activation can protect enteric neurons from damage due to diabetes in the acute stage of the disease, in part via the promotion of neurotrophin release.
Collapse
Affiliation(s)
- P Luo
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - D Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - C Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - W-X He
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - C-L Zhang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - M-J Chang
- Center for Translational Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Chen Y, Liu G, He F, Zhang L, Yang K, Yu H, Zhou J, Gan H. MicroRNA 375 modulates hyperglycemia-induced enteric glial cell apoptosis and Diabetes-induced gastrointestinal dysfunction by targeting Pdk1 and repressing PI3K/Akt pathway. Sci Rep 2018; 8:12681. [PMID: 30140011 PMCID: PMC6107553 DOI: 10.1038/s41598-018-30714-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Diabetic neuropathy can damage systemic nervous system, including alteration of enteric nervous system and subsequent gastrointestinal dysfunction. The effect of diabetes on enteric glia cell (EGC) is not clear. We investigated the effect of diabetes and hyperglycemia on EGC, and the role of microRNA375 in modulating EGC survival in vivo and in vitro. Streptozotocin-induced diabetic mice were intraperitoneally injected with microRNA375 inhibitor or its negative control. EGC was transfected with microRNA375 inhibitor or its mimic. Diabetes mice with gastrointestinal dysfunction showed increased apoptosis of EGC (no difference in cell numbers) and gene expression of micorRNA375 in the myenteric plexus. Hyperglycemia triggered apoptosis of EGC in vitro with decreased expression of Pdk1 and p-Akt, but increased expression of micorRNA375. MicorRNA375 mimic induced apoptosis of EGC in vitro with repressed Pdk1and p-Akt. MicorRNA375 inhibitor could both prevent hyperglycemia-induced apoptosis of EGC in vitro and diabetes-induced gastrointestinal dysfunction in vivo. Our results suggest that diabetes-induced gastrointestinal dysfunction is related to increased apoptosis of EGC in the myenteric plexus. Hyperglycemia can increase the expression of microRNA375 and damage EGC survival through PI3K/Akt pathway. MicroRNA375 specific inhibition can prevent hyperglycemia induced EGC damage and diabetes-induced gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Yan Chen
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gongxiang Liu
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fuqian He
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Zhang
- Department of elderly digestive, Sichuan Provincial People's Hospital, Chengdu, 610072, China
| | - Kun Yang
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huan Yu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinqiu Zhou
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huatian Gan
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
15
|
Sha H, Tong X, Zhao J. Abnormal expressions of AGEs, TGF-β1, BDNF and their receptors in diabetic rat colon-Associations with colonic morphometric and biomechanical remodeling. Sci Rep 2018; 8:9437. [PMID: 29930382 PMCID: PMC6013484 DOI: 10.1038/s41598-018-27787-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/08/2018] [Indexed: 02/06/2023] Open
Abstract
Present study aims to investigate the role of AGEs, TGF-β1, BDNF and their receptors on diabetes-induced colon remodeling. Diabetes was induced by a single tail vein injection 40 mg/kg of STZ. The parameters of morphometric and biomechanical properties of colonic segments were obtained from diabetic and normal rats. The expressions of AGE, RAGE, TGF- β1, TGF- β1 receptor, BDNF and TrkB were immunohistochemically detected in different layers of the colon. The expressions of AGE, RAGE, TGF-β1 and TGF- β1 receptor were increased whereas BDNF and TrkB were decreased in the diabetic colon (P < 0.05, P < 0.01). AGE, RAGE and TGF-β1 receptor expressions were positively correlated whereas the BDNF expression was negatively correlated with most of the morphometry and biomechanical parameters (P < 0.05, P < 0.01, P < 0.001). AGE, TGF- β1 and BDNF in different layers correlated with their receptors RAGE, TGF- β1 receptor and TrkB respectively. STZ-induced diabetes up-regulated the expression of AGE, RAGE, TGF- β1 and TGF- β1 receptors and down-regulated BDNF and TrkB in different layers of diabetic colon mainly due to hyperglycemia. Such changes maybe important for diabetes-induced colon remodeling, however it is needed to further perform mechanistic experiments in order to study causality or approaches that explain the relevance of the molecular pathways.
Collapse
Affiliation(s)
- Hong Sha
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Xiaolin Tong
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jingbo Zhao
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark.
| |
Collapse
|
16
|
Resveratrol promotes neuroprotection and attenuates oxidative and nitrosative stress in the small intestine in diabetic rats. Biomed Pharmacother 2018; 105:724-733. [PMID: 29906751 DOI: 10.1016/j.biopha.2018.06.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
Damages to the enteric nervous system caused by diabetes mellitus (DM) are frequently attributed to oxidative and nitrosative stress. We aimed to investigate the effect of Resveratrol (RSV) (10 mg/kg) on oxidative and nitrosative stress in the intestinal wall and morphoquantitative aspects of the myenteric plexus of the duodenum, jejunum and ileum in diabetic rats. Twenty-four rats were distributed into four groups (n = 6/group): control (C group), control treated with RSV (CR group), diabetic (D group), and diabetic treated with RSV (DR group) for 120 days. Immunohistochemical staining techniques for the general neuronal population, nitrergic and calretinin neuronal subpopulations, enteric glial cells and glial fibrillary acid protein were performed in the myenteric plexus. Furthermore, parameters of oxidative and nitrosative stress were analyzed in the intestinal wall. RSV attenuated oxidative and nitrosative stress and prevented neuronal loss and hypertrophy of the HuC/D-IR, nNOS-IR and CALR-IR neuronal subpopulations in the DR group compared with the D group (P < 0.05). In addition, RSV prevented the increase in glial fibrillary acid protein fluorescence in the DR group compared with the D (P < 0.05). These results suggest that RSV has antioxidant and neuroprotective effects in myenteric plexus in rats with experimental DM.
Collapse
|
17
|
Stojanovska V, McQuade RM, Miller S, Nurgali K. Effects of Oxaliplatin Treatment on the Myenteric Plexus Innervation and Glia in the Murine Distal Colon. J Histochem Cytochem 2018; 66:723-736. [PMID: 29741434 DOI: 10.1369/0022155418774755] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oxaliplatin (platinum-based chemotherapeutic agent) is a first-line treatment of colorectal malignancies; its use associates with peripheral neuropathies and gastrointestinal side effects. These gastrointestinal dysfunctions might be due to toxic effects of oxaliplatin on the intestinal innervation and glia. Male Balb/c mice received intraperitoneal injections of sterile water or oxaliplatin (3 mg/kg/d) triweekly for 2 weeks. Colon tissues were collected for immunohistochemical assessment at day 14. The density of sensory, adrenergic, and cholinergic nerve fibers labeled with calcitonin gene-related peptide (CGRP), tyrosine hydroxylase (TH), and vesicular acetylcholine transporter (VAChT), respectively, was assessed within the myenteric plexus of the distal colon. The number and proportion of excitatory neurons immunoreactive (IR) against choline acetyltransferase (ChAT) were counted, and the density of glial subpopulations was determined by using antibodies specific for glial fibrillary acidic protein (GFAP) and s100β protein. Oxaliplatin treatment induced significant reduction of sensory and adrenergic innervations, as well as the total number and proportion of ChAT-IR neurons, and GFAP-IR glia, but increased s100β expression within the myenteric plexus of the distal colon. Treatment with oxaliplatin significantly alters nerve fibers and glial cells in the colonic myenteric plexus, which could contribute to long-term gastrointestinal side effects following chemotherapeutic treatment.
Collapse
Affiliation(s)
- Vanesa Stojanovska
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Rachel M McQuade
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Sarah Miller
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia
| | - Kulmira Nurgali
- College of Health and Biomedicine, Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia.,Department of Medicine Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Regenerative Medicine and Stem Cells Program, Australian Institute for Musculoskeletal Science, Melbourne, Victoria, Australia
| |
Collapse
|
18
|
Vicentino-Vieira SL, Góis MB, Trevizan AR, de Lima LL, Leatte EP, Nogueira de Melo GDA, Garcia JL, Araújo EJDA, Sant'Ana DDMG. Toxoplasma gondii infection causes structural changes in the jejunum of rats infected with different inoculum doses. Life Sci 2017; 191:141-149. [DOI: 10.1016/j.lfs.2017.10.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/04/2017] [Accepted: 10/23/2017] [Indexed: 01/08/2023]
|
19
|
Fukuhara S, Masaoka T, Nishimura S, Nakamura M, Matsuzaki J, Tsugawa H, Miyoshi S, Mori H, Kawase S, Shibata S, Okano H, Kanai T, Suzuki H. Enteric Glial Dysfunction Evoked by Apolipoprotein E Deficiency Contributes to Delayed Gastric Emptying. Dig Dis Sci 2017; 62:3359-3369. [PMID: 29098550 DOI: 10.1007/s10620-017-4820-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 10/20/2017] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIM Diabetes is the main cause of gastroparesis accompanying decreased neuronal nitric oxide synthase (nNOS) in myenteric ganglia of the stomach. Decreased nNOS expression in the stomach also results from defects in apolipoprotein E (ApoE), which is secreted by astrocytes and has neuroprotective effects on the central nervous system. However, the roles of ApoE and enteric glial cells on gastric motility are uncertain. In this study, ApoE and enteric glial cell alterations in gastroparesis were investigated. METHODS Type 2 diabetic (db/db) mice and ApoE-knockout mice were analyzed. Gastric emptying was measured using the 13C acetic acid breath test. Expression levels of the pan-neuronal marker, protein gene product 9.5 (PGP 9.5), and glial marker, glial fibrillary acidic protein (GFAP) were examined by immunohistochemistry. Neural stem cells (NSCs) were injected into the gastric antral wall of ApoE-knockout mice. RESULTS Delayed gastric emptying was observed in 27% of db/db mice with significant decreases in serum ApoE levels and GFAP expression in the gastric antrum. Gastric emptying was also delayed in ApoE-knockout mice, with a significant decrease in GFAP expression, but no change in PGP 9.5 expression. Transplantation of NSCs improved gastric emptying in ApoE-knockout mice through supplementation of GFAP-positive cells. CONCLUSIONS Our results suggest that decreased enteric glial cells in ApoE-knockout mice are crucial for development of delayed gastric emptying, and NSC transplantation is effective in restoring myenteric ganglia and gastric motility.
Collapse
Affiliation(s)
- Seiichiro Fukuhara
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Tatsuhiro Masaoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Soraya Nishimura
- Department of Orthopedics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopedics, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Juntaro Matsuzaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Hitoshi Tsugawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Sawako Miyoshi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Hideki Mori
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Satoshi Kawase
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Hidekazu Suzuki
- Medical Education Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
20
|
Robinson AM, Stojanovska V, Rahman AA, McQuade RM, Senior PV, Nurgali K. Effects of Oxaliplatin Treatment on the Enteric Glial Cells and Neurons in the Mouse Ileum. J Histochem Cytochem 2016; 64:530-45. [PMID: 27389702 PMCID: PMC5006136 DOI: 10.1369/0022155416656842] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/06/2016] [Indexed: 12/17/2022] Open
Abstract
Oxaliplatin, currently used for treatment of colorectal and other cancers, causes severe gastrointestinal side effects, including nausea, vomiting, diarrhea, and constipation that are attributed to mucosal damage. However, delayed onset and long-term persistence of these side effects suggest that damage to the enteric nervous system (ENS) regulating physiological function of the gastrointestinal tract may also occur. The ENS comprises myenteric and submucosal neurons and enteric glial cells (EGCs). This study aimed to investigate the effects of oxaliplatin treatment on enteric neurons and EGCs within the mouse ileum. BALB/c mice received repeated intraperitoneal injections of oxaliplatin (3 mg/kg, 3 injections/week). Tissues were collected 3, 7, 14, and 21 days from the commencement of treatment. Decreases in glial fibrillary acidic protein-immunoreactive (IR) EGCs and protein gene product 9.5/β-Tubulin III-IR neurons as well as increase in s100β-IR EGCs after chronic oxaliplatin administration were observed in both the myenteric and submucosal plexi. Changes in EGCs were further observed in cross-sections of the ileum at day 14 and confirmed by Western blotting. Alterations in EGCs correlated with loss of myenteric and submucosal neurons in the ileum from oxaliplatin-treated mice. These changes to the ENS may contribute to the mechanisms underlying gastrointestinal side effects associated with oxaliplatin treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Kulmira Nurgali
- Kulmira Nurgali, Western Centre for Health Research & Education, Sunshine Hospital, 176 Furlong Road, St Albans, VIC 3021, Australia.
| |
Collapse
|
21
|
Cui W, Zhang Y, Lu D, Ren M, Yuan G. Upregulation of p‑Akt by glial cell line‑derived neurotrophic factor ameliorates cell apoptosis in the hippocampus of rats with streptozotocin‑induced diabetic encephalopathy. Mol Med Rep 2015; 13:543-9. [PMID: 26549420 DOI: 10.3892/mmr.2015.4507] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 09/22/2015] [Indexed: 11/06/2022] Open
Abstract
The loss of neurotrophic factor support has been shown to contribute to the development of the central nervous system. Glial cell line‑derived neurotrophic factor (GDNF), a potent neurotrophic factor, is closely associated with apoptosis and exerts neuroprotective effects on numerous populations of cells. However, the underlying mechanisms of these protective effects remain unknown. In the present study, a significant increase in Bax levels and DNA fragmentation was observed in the hippocampus obtained from the brains of diabetic rats 60 days after diabetes had been induced. The apoptotic changes were correlated with the loss of GDNF/Akt signaling. GDNF administration was found to reverse the diabetes‑induced Bax and DNA fragmentation changes. This was associated with an improvement in the level of p‑Akt/Akt. In addition, combination of GDNF with a specific inhibitor of the phosphoinositide 3‑kinase (PI3K)/Akt pathway, Wortmannin, significantly abrogated the effects of GDNF on the levels of p‑Akt/Akt, Bax and DNA fragmentation. However, a p38 mitogen‑activated proten kinase (MAPK) inhibitor, SB203580, had no effect on the expression of p‑Akt/Akt, Bax or DNA fragmentation. These results demonstrate the pivotal role of GDNF as well as the PI3K/Akt pathway, but not the MAPK pathway, in the prevention of diabetes‑induced neuronal apoptosis in the hippocampus.
Collapse
Affiliation(s)
- Weigang Cui
- Henan Key Laboratory of Medical Tissue Regeneration, Department of Human Anatomy, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Yinghua Zhang
- Henan Key Laboratory of Medical Tissue Regeneration, Department of Human Anatomy, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Derong Lu
- Department of Internal Digestive Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Mingxin Ren
- Henan Key Laboratory of Medical Tissue Regeneration, Department of Human Anatomy, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Guoyan Yuan
- Department of Neurosurgery, The First Affiliated Hospital, Xinxiang Medical University, Weihui, Henan 453100, P.R. China
| |
Collapse
|
22
|
de Oliveira Belém M, Cirilo CP, de Santi-Rampazzo AP, Schoffen JPF, Comar JF, Natali MRM, de Almeida Araújo EJ. Intestinal morphology adjustments caused by dietary restriction improves the nutritional status during the aging process of rats. Exp Gerontol 2015; 69:85-93. [DOI: 10.1016/j.exger.2015.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 05/05/2015] [Accepted: 05/06/2015] [Indexed: 12/27/2022]
|
23
|
Aqueous Extract of Agaricus blazei Murrill Prevents Age-Related Changes in the Myenteric Plexus of the Jejunum in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:287153. [PMID: 25960748 PMCID: PMC4415631 DOI: 10.1155/2015/287153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/27/2023]
Abstract
This study evaluated the effects of the supplementation with aqueous extract of Agaricus blazei Murrill (ABM) on biometric and blood parameters and quantitative morphology of the myenteric plexus and jejunal wall in aging Wistar rats. The animals were euthanized at 7 (C7), 12 (C12 and CA12), and 23 months of age (C23 and CA23). The CA12 and CA23 groups received a daily dose of ABM extract (26 mg/animal) via gavage, beginning at 7 months of age. A reduction in food intake was observed with aging, with increases in the Lee index, retroperitoneal fat, intestinal length, and levels of total cholesterol and total proteins. Aging led to a reduction of the total wall thickness, mucosa tunic, villus height, crypt depth, and number of goblet cells. In the myenteric plexus, aging quantitatively decreased the population of HuC/D(+) neuronal and S100(+) glial cells, with maintenance of the nNOS(+) nitrergic subpopulation and increase in the cell body area of these populations. Supplementation with the ABM extract preserved the myenteric plexus in old animals, in which no differences were detected in the density and cell body profile of neurons and glial cells in the CA12 and CA23 groups, compared with C7 group. The supplementation with the aqueous extract of ABM efficiently maintained myenteric plexus homeostasis, which positively influenced the physiology and prevented the death of the neurons and glial cells.
Collapse
|
24
|
Zhang GQ, Yang S, Li XS, Zhou DS. Expression and possible role of IGF-IR in the mouse gastric myenteric plexus and smooth muscles. Acta Histochem 2014; 116:788-94. [PMID: 24630395 DOI: 10.1016/j.acthis.2014.01.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/10/2014] [Accepted: 01/13/2014] [Indexed: 12/26/2022]
Abstract
Insulin-like growth factor-I (IGF-I) and its receptor (IGF-IR) have tremendous trophic effects on the central, peripheral and enteric neurons. The loss of IGF-IR contributes to the development of diabetic gastroparesis. However, the nature and the function of the IGF-IR(+) cells in the gastric myenteric plexus remain unclear. In this study, anti-ChAT, anti-S100β or anti-c-KIT antibodies were used to co-label IGF-IR(+) cells and neurons, glial cells or interstitial cells of Cajal (ICCs), respectively. We also generated type 1 diabetic mice (DM) to explore the influence of impaired IGF-I/IGF-IR in the myenteric neurons. Results showed that IGF-IR was expressed in the epithelium, smooth muscles and myenteric plexi of the mouse stomach. Most of the IGF-IR(+) cells in the myenteric plexi were ChAT(+) cholinergic neurons, but not enteric glial cells and there were more IGF-IR(+) neurons and fibers in the gastric antrum than in the corpus. The IGF-IR(+)/ChAT(+) neurons and ICCs were closely juxtaposed, but distinctly distributed in the myenteric plexus, indicating a possible role for the IGF-IR(+)/ChAT(+) neurons in the mediation of gastric motility through ICCs. Moreover, the decrease of IGF-IR and cholinergic neurons in the myenteric plexi and smooth muscles of DM mice suggested that IGF-I/IGF-IR signaling might play a role in neuron survival and neurite outgrowth, as well as stem cell factor (SCF) production, which is required for the development of ICCs. Our results provide insights into the effects of IGF-I/IGF-IR signaling on the development of gastrointestinal motility disorders.
Collapse
|
25
|
Schoffen JPF, Santi Rampazzo AP, Cirilo CP, Zapater MCU, Vicentini FA, Comar JF, Bracht A, Natali MRM. Food restriction enhances oxidative status in aging rats with neuroprotective effects on myenteric neuron populations in the proximal colon. Exp Gerontol 2014; 51:54-64. [DOI: 10.1016/j.exger.2014.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 01/26/2023]
|
26
|
Abstract
Autonomic neuropathy complicates diabetes by increasing patient morbidity and mortality. Surprisingly, considering its importance, development and exploitation of animal models has lagged behind the wealth of information collected for somatic symmetrical sensory neuropathy. Nonetheless, animal studies have resulted in a variety of insights into the pathogenesis, neuropathology, and pathophysiology of diabetic autonomic neuropathy (DAN) with significant and, in some cases, remarkable correspondence between rodent models and human disease. Particularly in the study of alimentary dysfunction, findings in intrinsic intramural ganglia, interstitial cells of Cajal and the extrinsic parasympathetic and sympathetic ganglia serving the bowel vie for recognition as the chief mechanism. A body of work focused on neuropathologic findings in experimental animals and human subjects has demonstrated that axonal and dendritic pathology in sympathetic ganglia with relative neuron preservation represents one of the neuropathologic hallmarks of DAN but it is unlikely to represent the entire story. There is a surprising selectivity of the diabetic process for subpopulations of neurons and nerve terminals within intramural, parasympathetic, and sympathetic ganglia and innervation of end organs, afflicting some while sparing others, and differing between vascular and other targets within individual end organs. Rather than resulting from a simple deficit in one limb of an effector pathway, autonomic dysfunction may proceed from the inability to integrate portions of several complex pathways. The selectivity of the diabetic process appears to confound a simple global explanation (e.g., ischemia) of DAN. Although the search for a single unifying pathogenetic hypothesis continues, it is possible that autonomic neuropathy will have multiple pathogenetic mechanisms whose interplay may require therapies consisting of a cocktail of drugs. The role of multiple neurotrophic substances, antioxidants (general or pathway specific), inhibitors of formation of advanced glycosylation end products and drugs affecting the polyol pathway may be complex and therapeutic elements may have both salutary and untoward effects. This review has attempted to present the background and current findings and hypotheses, focusing on autonomic elements including and beyond the typical parasympathetic and sympathetic nervous systems to include visceral sensory and enteric nervous systems.
Collapse
Affiliation(s)
- Robert E Schmidt
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
27
|
Brun P, Giron MC, Qesari M, Porzionato A, Caputi V, Zoppellaro C, Banzato S, Grillo AR, Spagnol L, De Caro R, Pizzuti D, Barbieri V, Rosato A, Sturniolo GC, Martines D, Zaninotto G, Palù G, Castagliuolo I. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 2013; 145:1323-33. [PMID: 23994200 DOI: 10.1053/j.gastro.2013.08.047] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 08/03/2013] [Accepted: 08/15/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS In the intestines, Toll-like receptor 2 (TLR2) mediates immune responses to pathogens and regulates epithelial barrier function; polymorphisms in TLR2 have been associated with inflammatory bowel disease phenotype. We assessed the effects of TLR2 signaling on the enteric nervous system (ENS) in mice. METHODS TLR2 distribution and function in the ileal neuromuscular layer of mice were determined by immunofluorescence, cytofluorimetric analysis, immunoprecipitation, and immunoblot analyses. We assessed morphology and function of the ENS in Tlr2(-/-) mice and in mice with wild-type Tlr2 (wild-type mice) depleted of intestinal microbiota, using immunofluorescence, immunoblot, and gastrointestinal motility assays. Levels and signaling of glial cell line-derived neurotrophic factor (GDNF) were determined using quantitative reverse transcriptase polymerase chain reaction, immunohistochemistry, and immunoprecipitation analyses. Colitis was induced by administration of dextran sulfate sodium or 2,4 dinitrobenzensulfonic acid to Tlr2(-/-) mice after termination of GDNF administration. RESULTS TLR2 was expressed in enteric neurons, glia, and smooth muscle cells of the intestinal wall. Tlr2(-/-) mice had alterations in ENS architecture and neurochemical profile, intestinal dysmotility, abnormal mucosal secretion, reduced levels of GDNF in smooth muscle cells, and impaired signaling via Ret-GFRα1. ENS structural and functional anomalies were completely corrected by administration of GDNF to Tlr2(-/-) mice. Wild-type mice depleted of intestinal microbiota had ENS defects and GDNF deficiency, similar to Tlr2(-/-) mice; these defects were partially restored by administration of a TLR2 agonist. Tlr2(-/-) mice developed more severe colitis than wild-type mice after administration of dextran sulfate sodium or 2,4 dinitrobenzensulfonic acid; colitis was not more severe if Tlr2(-/-) mice were given GDNF before dextran sulfate sodium or 2,4 dinitrobenzensulfonic acid. CONCLUSIONS In mice, TLR2 signaling regulates intestinal inflammation by controlling ENS structure and neurochemical coding, along with intestinal neuromuscular function. These findings provide information as to how defective TLR2 signaling in the ENS affects inflammatory bowel disease phenotype in humans.
Collapse
Affiliation(s)
- Paola Brun
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Stenkamp-Strahm C, Patterson S, Boren J, Gericke M, Balemba O. High-fat diet and age-dependent effects on enteric glial cell populations of mouse small intestine. Auton Neurosci 2013; 177:199-210. [PMID: 23726157 DOI: 10.1016/j.autneu.2013.04.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/26/2013] [Accepted: 04/29/2013] [Indexed: 02/09/2023]
Abstract
Diabetes and obesity are increasing in prevalence at an alarming rate throughout the world. Autonomic diabetic neuropathy is evident in individuals that experience a long-standing diabetic disease state, and gastrointestinal (GI) dysmotility is thought to be the outcome of neuropathies within the enteric nervous system (ENS) of these patients. To date, an analysis of enteric glial cell population changes during diabetic symptoms has not been performed, and may bring insight into disease pathology and neuropathy, given glial cell implications in gastrointestinal and neuronal homeostasis. Diabetes and obesity were monitored in C57Bl/6J mice fed a 72% high-fat diet, and duodenal glial expression patterns were evaluated by immunohistochemistry and RT-PCR for S100β, Sox10 and GFAP proteins and transcripts, as well as transmission electron microscopy (TEM). The high-fat diet caused obesity, hyperglycemia and insulin resistance after 4 weeks. These changes were associated with a significant decline in the area density indices of mucosa-associated glial cell networks, evidenced by S100β staining at 8 and 20 weeks. All three markers and TEM showed that myenteric glial cells were unaffected by early and late disease periods. However, analysis of Sox10 transcript expression and immunoreactivity showed a diet independent, age-associated decline in glial cell populations. This is the first study showing that mucosal glia cell damage occurs during diabetic symptoms, suggesting that mucosal enteric glia injury may have a pathophysiological significance during this disease. Our results also provide support for age-associated changes in longitudinal studies of enteric glial cells.
Collapse
|
29
|
LePard KJ, Cellini J. Age-dependent slowing of enteric axonal transport in insulin-resistant mice. World J Gastroenterol 2013; 19:482-91. [PMID: 23382626 PMCID: PMC3558571 DOI: 10.3748/wjg.v19.i4.482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 11/20/2012] [Accepted: 12/15/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate retrograde tracer transport by gastric enteric neurons in insulin resistant mice with low or high glycosylated hemoglobin (Hb).
METHODS: Under anesthesia, the retrograde tracer fluorogold was superficially injected into the fundus or antrum using a microsyringe in KK Cg-Ay/J mice prior to onset of type 2 diabetes mellitus (T2DM; 4 wk of age), at onset of T2DM (8 wk of age), and after 8, 16, or 24 wk of untreated T2DM and in age-matched KK/HIJ mice. Six days later, mice were sacrificed by CO2 narcosis followed by pneumothorax. Stomachs were removed and fixed. Sections from fundus, corpus and antrum were excised and mounted on a glass slide. Tracer-labeled neurons were viewed using a microscope and manually counted. Data were expressed as the number of neurons in short and long descending and ascending pathways and in local fundus and antrum pathways, and the number of neurons in all regions labeled after injection of tracer into either the fundus or the antrum.
RESULTS: By 8 wk of age, body weights of KKAy mice (n = 12, 34 ± 1 g) were heavier than KK mice (n = 17, 29 ± 1 g; F (4, 120) = 4.414, P = 0.002] and glycosylated Hb was higher [KK: (n = 7), 4.97% ± 0.04%; KKAy: (n = 6), 6.57% ± 0.47%; F (1, 26) = 24.748, P < 0.001]. The number of tracer labeled enteric neurons was similar in KK and KKAy mice of all ages in the short descending pathway [F (1, 57) = 2.374, P = 0.129], long descending pathway [F (1, 57) = 0.922, P = 0.341], local fundus pathway [F (1, 53) = 2.464, P = 0.122], local antrum pathway [F (1, 57) = 0.728, P = 0.397], and short ascending pathway [F (1, 53) = 2.940, P = 0.092]. In the long ascending pathway, fewer tracer-labeled neurons were present in KKAy as compared to KK mice [KK: (n = 34), 302 ± 17; KKAy: (n = 29), 230 ± 15; F (1, 53) = 8.136, P = 0.006]. The number of tracer-labeled neurons was decreased in all mice by 16 wk as compared to 8 wk of age in the short descending pathway [8 wk: (n = 15), 305 ± 26; 16 wk: (n = 13), 210 ± 30; F (4, 57) = 9.336, P < 0.001], local antrum pathway [8 wk: (n = 15), 349 ± 20; 16 wk: (n = 13), 220 ± 33; F (4, 57) = 8.920, P < 0.001], short ascending pathway [8 wk: (n = 14), 392 ± 15; 16 wk: (n = 14), 257 ± 33; F (4, 53) = 17.188, P < 0.001], and long ascending pathway [8 wk: (n = 14), 379 ± 39; 16 wk: (n = 14), 235 ± 26; F (4, 53) = 24.936, P < 0.001]. The number of tracer-labeled neurons decreased at 24 wk of age in the local fundus pathway [8 wk: (n = 14), 33 ± 11; 24 wk: (n = 12), 3 ± 2; F (4, 53) = 5.195, P = 0.001] and 32 wk of age in the long descending pathway [8 wk: (n = 15), 16 ± 3; 32 wk: (n = 12), 3 ± 2; F (4, 57) = 2.944, P = 0.028]. The number of tracer-labeled enteric neurons was correlated to final body weight for local fundus and ascending pathways [KK: (n = 34), r = -0.746, P < 0.001; KKAy: (n = 29), r = -0.842, P < 0.001] as well as local antrum and descending pathways [KK (n = 36), r = -0.660, P < 0.001; KKAy (n = 31), r = -0.622, P < 0.001]. In contrast, glycosylated Hb was not significantly correlated to number of tracer-labeled neurons [KK (n = 17), r = -0.164, P = 0.528; KKAy (n = 16), r = -0.078, P = 0.774].
CONCLUSION: Since uncontrolled T2DM did not uniformly impair tracer transport in gastric neurons, long ascending neurons may be more susceptible to persistent hyperglycemia and low effective insulin.
Collapse
|
30
|
Dietary restriction interferes with oxidative status and intrinsic intestinal innervation in aging rats. Nutrition 2013; 29:673-80. [PMID: 23317927 DOI: 10.1016/j.nut.2012.09.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/20/2012] [Accepted: 09/24/2012] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To evaluate the effects of dietary restriction on oxidative status, the HuC/D-neuronal nitric oxide synthase (nNOS) myenteric neuron population, HuC/D-S100 glial cells, and the morphometry of the small intestine in rats at various ages. METHODS Fifteen Wistar rats were divided into 7-and 12-mo-old control groups and a 12-mo-old experimental group subjected to dietary restrictions (50% of normal ration) for 5 mo. At 7 and 12 mo of age, the animals were anesthetized, and blood was collected to assess the biochemical components and oxidative status. Ileum samples were subjected to double-marker (HuC/D-nNOS and HuC/D-S100) immunostaining and histologic processing to morphometrically analyze intestinal wall elements and determine the metaphase index and rate of caliciform cells. The data were subjected to analysis of variance and the Tukey post hoc test with a 5% significance level. RESULTS Age affected the oxidative status by increasing lipid peroxidation, with no effect on blood components, intrinsic innervation, and intestinal wall elements. The animals subjected to dietary restriction showed improved levels of total cholesterol, triacylglycerols, and oxidative status, with no changes in the nNOS neuron population. However, the dietary restriction dramatically decreased the glial and HuC/D myenteric populations, led to atrophy of the neuronal cell body, induced glial hypertrophy, and decreased the thickness of the intestinal wall. CONCLUSION The high oxidative status of the aging animals was reversed by dietary restriction, which also lowered cholesterol and triacylglycerol levels. The present dietary restriction elicited morpho-quantitative changes in the myenteric plexus and histology of the ileum, with likely effects on intestinal functions.
Collapse
|
31
|
Bagyánszki M, Bódi N. Diabetes-related alterations in the enteric nervous system and its microenvironment. World J Diabetes 2012; 3:80-93. [PMID: 22645637 PMCID: PMC3360223 DOI: 10.4239/wjd.v3.i5.80] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 04/06/2012] [Accepted: 05/11/2012] [Indexed: 02/05/2023] Open
Abstract
Gastric intestinal symptoms common among diabetic patients are often caused by intestinal motility abnormalities related to enteric neuropathy. It has recently been demonstrated that the nitrergic subpopulation of myenteric neurons are especially susceptible to the development of diabetic neuropathy. Additionally, different susceptibility of nitrergic neurons located in different intestinal segments to diabetic damage and their different levels of responsiveness to insulin treatment have been revealed. These findings indicate the importance of the neuronal microenvironment in the pathogenesis of diabetic nitrergic neuropathy. The main focus of this review therefore was to summarize recent advances related to the diabetes-related selective nitrergic neuropathy and associated motility disturbances. Special attention was given to the findings on capillary endothelium and enteric glial cells. Growing evidence indicates that capillary endothelium adjacent to the myenteric ganglia and enteric glial cells surrounding them are determinative in establishing the ganglionic microenvironment. Additionally, recent advances in the development of new strategies to improve glycemic control in type 1 and type 2 diabetes mellitus are also considered in this review. Finally, looking to the future, the recent and promising results of metagenomics for the characterization of the gut microbiome in health and disease such as diabetes are highlighted.
Collapse
Affiliation(s)
- Mária Bagyánszki
- Mária Bagyánszki, Nikolett Bódi, Department of Physiology, Anatomy and Neuroscience, Faculty of Science, University of Szeged, H-6726 Szeged, Hungary
| | | |
Collapse
|
32
|
Korsak K, Silva AT, Saffrey MJ. Differing effects of NT-3 and GDNF on dissociated enteric ganglion cells exposed to hydrogen peroxide in vitro. Neurosci Lett 2012; 517:102-6. [PMID: 22548772 DOI: 10.1016/j.neulet.2012.04.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 03/26/2012] [Accepted: 04/13/2012] [Indexed: 01/19/2023]
Abstract
Oxidative stress is widely recognized to contribute to neuronal death during various pathological conditions and ageing. In the enteric nervous system (ENS), reactive oxygen species have been implicated in the mechanism of age-associated neuronal loss. The neurotrophic factors, neurotrophin 3 (NT-3) and glial cell line-derived neurotrophic factor (GDNF), are important in the development of enteric neurons and continue to be expressed in the gut throughout life. It has therefore been suggested that they may have a neuroprotective role in the ENS. We investigated the potential of NT-3 and GDNF to prevent the death of enteric ganglion cells in dissociated cell culture after exposure to hydrogen peroxide (H(2)O(2)). H(2)O(2) treatment resulted in a dose-dependent death of enteric neurons and glial cells, as demonstrated by MTS assay, bis-benzimide and propidium iodide staining and immunolabelling. Cultures treated with NT-3 prior to exposure showed reduced cell death compared to untreated control or GDNF-treated cultures. GDNF treatment did not affect neuronal survival in H(2)O(2)-treated cultures. These results suggest that NT-3 is able to enhance the survival of enteric ganglion cells exposed to oxidative stress.
Collapse
Affiliation(s)
- Kris Korsak
- Department of Life, Health and Chemical Sciences, The Open University, UK
| | | | | |
Collapse
|
33
|
Pereira RVF, Tronchini EA, Tashima CM, Alves EPB, Lima MM, Zanoni JN. L-glutamine supplementation prevents myenteric neuron loss and has gliatrophic effects in the ileum of diabetic rats. Dig Dis Sci 2011; 56:3507-16. [PMID: 21710226 DOI: 10.1007/s10620-011-1806-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 06/14/2011] [Indexed: 12/16/2022]
Abstract
BACKGROUND Peripheral neuropathy caused chronically by diabetes mellitus is related to exacerbation of oxidative stress and a significant reduction in important endogenous antioxidants. L: -Glutamine is an amino acid involved in defense mechanisms and is a substrate for the formation of glutathione, the major endogenous cellular antioxidant. AIM This study investigated the effects of 2% L: -glutamine supplementation on peripheral diabetic neuropathy and enteric glia in the ileum in rats. METHODS Male Wistar rats were divided into four groups: normoglycemics (N), normoglycemics supplemented with L: -glutamine (NG), diabetics (D), and diabetics supplemented with L: -glutamine (DG). After 120 days, the ileums were processed for HuC/D and S100 immunohistochemistry. Quantitative and morphometric analysis was performed. RESULTS Diabetes significantly reduced the number of HuC/D-immunoreactive myenteric neurons per unit area and per ganglion in group D compared with normoglycemic animals (group N). L: -Glutamine (2%) prevented neuronal death induced by diabetes (group DG) compared with group D. The glial density per unit area did not change with diabetes (group D) but was significantly reduced after L: -glutamine supplementation (groups NG and DG). Ganglionic glial density was similar among the four groups. The neuronal area was not altered in groups D and DG. Glial size was reduced in group D; this was reversed by L: -glutamine supplementation (group DG). CONCLUSIONS We concluded that 2% L: -glutamine had neuroprotective effects directly on myenteric neurons and indirectly through glial cells, which had gliatrophic effects.
Collapse
Affiliation(s)
- Renata Virginia Fernandes Pereira
- Department of Morphological Sciences, Universidade Estadual de Maringá, Avenida Colombo, nº 5790 Bloco H-79, Maringá, PR, CEP 87020-900, Brazil.
| | | | | | | | | | | |
Collapse
|
34
|
Domènech A, Pasquinelli G, De Giorgio R, Gori A, Bosch F, Pumarola M, Jiménez M. Morphofunctional changes underlying intestinal dysmotility in diabetic RIP-I/hIFNβ transgenic mice. Int J Exp Pathol 2011; 92:400-12. [PMID: 22050417 DOI: 10.1111/j.1365-2613.2011.00789.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The pathogenetic mechanisms underlying gastrointestinal dysmotility in diabetic patients remain poorly understood, although enteric neuropathy, damage to interstitial cells of Cajal (ICC) and smooth muscle cell injury are believed to play a role. The aim of this study was to investigate the morphological and functional changes underlying intestinal dysmotility in RIP-I/hIFNβ transgenic mice treated with multiple very low doses of streptozotocin (20 mg/kg, i.p., 5 days). Compared with vehicle-treated mice, streptozotocin-treated animals developed type 1 diabetes mellitus, with sustained hyperglycaemia for 3.5 months, polyphagia, polydipsia and increased faecal output without changes in faecal water content (metabolic cages). Diabetic mice had a longer intestine, longer ileal villi and wider colonic crypts (conventional microscopy) and displayed faster gastric emptying and intestinal transit. Contractility studies showed selective impaired neurotransmission in the ileum and mid-colon of diabetic mice. Compared with controls, the ileal and colonic myenteric plexus of diabetic mice revealed ultrastructural features of neuronal degeneration and HuD immunohistochemistry on whole-mount preparations showed 15% reduction in neuronal numbers. However, no immunohistochemical changes in apoptosis-related markers were noted. Lower absolute numbers of neuronal nitric oxide synthase- and choline acetyltransferase-immunopositive neurons and enhanced vasoactive intestinal polypeptide and substance P immunopositivity were observed. Ultrastructural and immunohistochemical analyses did not reveal changes in the enteric glial or ICC networks. In conclusion, this model of diabetic enteropathy shows enhanced intestinal transit associated with intestinal remodelling, including neuroplastic changes, and overt myenteric neuropathy. Such abnormalities are likely to reflect neuroadaptive and neuropathological changes occurring in this diabetic model.
Collapse
Affiliation(s)
- Anna Domènech
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | | | | | | | | | | | | |
Collapse
|
35
|
Thacker M, Rivera LR, Cho HJ, Furness JB. The relationship between glial distortion and neuronal changes following intestinal ischemia and reperfusion. Neurogastroenterol Motil 2011; 23:e500-9. [PMID: 21410600 DOI: 10.1111/j.1365-2982.2011.01696.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Damage to mucosal epithelial cells, muscle cells and enteric neurons has been extensively studied following intestinal ischemia and reperfusion (I/R). Interestingly, the effects of intestinal I/R on enteric glia remains unexplored, despite knowledge that glia contribute to neuronal maintenance. Here, we describe structural damage to enteric glia and associated changes in distribution and immunoreactivity of the neuronal protein Hu. METHODS The mouse small intestine was made ischemic for 3 h and reperfused from 1 to 12 h. Immunohistochemical localisation of glial fibrillary acidic protein (GFAP), Hu and TUNEL were used to evaluate changes. KEY RESULTS At all time points glial cells became distorted, which was evident by their altered GFAP immunoreactivity, including an unusual appearance of bright perinuclear GFAP staining and the presence of GFAP globules. The numbers of neurons per ganglion area were significantly fewer in ganglia that contained distorted glia when compared with ganglia that contained glia of normal appearance. The distribution of Hu immunoreactivity was altered at all reperfusion time points. The presence of vacuoles and Hu granules in neurons was evident and an increase in nuclear Hu, relative to cytoplasmic Hu, was observed in ganglia that contained both normal and distorted glial cells. A number of neurons appeared to lose their Hu immunoreactivity, most noticeably in ganglia that contained distorted glial cells. TUNEL reaction occurred in a minority of glial cells and neurons. CONCLUSIONS & INFERENCES Structural damage to gliofilaments occurs following I/R and may be associated with damage to neighboring neurons.
Collapse
Affiliation(s)
- M Thacker
- Department of Anatomy & Cell Biology, University of Melbourne, Parkville, Vic., Australia.
| | | | | | | |
Collapse
|
36
|
Neuronal loss and abnormal BMP/Smad signaling in the myenteric plexus of diabetic rats. Auton Neurosci 2011; 164:51-61. [PMID: 21737358 DOI: 10.1016/j.autneu.2011.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2010] [Revised: 06/08/2011] [Accepted: 06/09/2011] [Indexed: 12/25/2022]
Abstract
Bone morphogenetic proteins (BMPs) are critical molecules during gut morphogenesis. However, little is known about their participation in the homeostasis of adult gut and their possible role in diseases. Gastrointestinal complications occur during diabetes with loss of enteric neurons. In this study, we investigated the possible involvement of BMPs signaling pathway in diabetic enteric neuropathy in an experimental model of diabetes in rats. The expression of BMPs, BMPs receptors and intracellular Smad effectors were assessed in control and diabetic smooth muscle layer of jejunum by immunofluorescence, Western blot and RT-PCR methods. Myenteric neurons and glial cells were measured by immunofluorescence using specific markers. In addition, cell apoptosis was evaluated by means of direct and indirect techniques. We demonstrated that diabetic ganglia displayed a significant decrease in ganglion size due to enhanced apoptosis and loss of peripherin. A decrease in glial fibrillary acidic protein (GFAP protein) was also observed in enteric glial cells. BMP-2 was down-regulated in the myenteric plexus of diabetic rats at 3 and 9weeks. A loss of enteric neurons by apoptosis was correlated with an ectopic BMP-4, increased BMPR-Ia and nuclear p-Smad1 expression in the myenteric plexus. Insulin-treatment prevented the intestinal alterations observed. These findings suggest that diabetes is associated with an abnormal BMP/Smad signaling expression in the myenteric ganglia that affects the homeostasis of the enteric plexus.
Collapse
|
37
|
Ashrafian H, Athanasiou T, Li JV, Bueter M, Ahmed K, Nagpal K, Holmes E, Darzi A, Bloom SR. Diabetes resolution and hyperinsulinaemia after metabolic Roux-en-Y gastric bypass. Obes Rev 2011; 12:e257-72. [PMID: 20880129 DOI: 10.1111/j.1467-789x.2010.00802.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The global prevalence of type 2 diabetes mellitus and impaired glucose metabolism continues to rise in conjunction with the pandemic of obesity. The metabolic Roux-en-Y gastric bypass operation offers the successful resolution of diabetes in addition to sustained weight loss and excellent long-term outcomes in morbidly obese individuals. The procedure consists of the physiological BRAVE effects: (i) Bile flow alteration; (ii) Reduction of gastric size; (iii) Anatomical gut rearrangement and altered flow of nutrients; (iv) Vagal manipulation and (v) Enteric gut hormone modulation. This operation provides anti-diabetic effects through decreasing insulin resistance and increasing the efficiency of insulin secretion. These metabolic outcomes are achieved through weight-independent and weight-dependent mechanisms. These include the foregut, midgut and hindgut mechanisms, decreased inflammation, fat, adipokine and bile metabolism, metabolic modulation, shifts in gut microbial composition and intestinal gluconeogenesis. In a small minority of patients, gastric bypass results in hyperinsulinaemic hypoglycaemia that may lead to nesidioblastosis (pancreatic beta-cell hypertrophy with islet hyperplasia). Elucidating the precise metabolic mechanisms of diabetes resolution and hyperinsulinaemia after surgery can lead to improved operations and disease-specific procedures including 'diabetes surgery'. It can also improve our understanding of diabetes pathogenesis that may provide novel strategies for the management of metabolic syndrome and impaired glucose metabolism.
Collapse
Affiliation(s)
- H Ashrafian
- The Department of Surgery and Cancer, Imperial College London, London, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kleinschmidt S, Nolte I, Hewicker-Trautwein M. Structural and functional changes of neuronal and glial components of the feline enteric nervous system in cats with chronic inflammatory and non-inflammatory diseases of the gastrointestinal tract. Res Vet Sci 2011; 91:e129-35. [PMID: 21349562 DOI: 10.1016/j.rvsc.2011.01.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 01/18/2011] [Accepted: 01/20/2011] [Indexed: 12/12/2022]
Abstract
Immunohistochemical examinations of the enteric nervous system (ENS) were performed on biopsies of healthy cats and compared to findings in cats suffering from inflammatory bowel disease or intestinal lymphoma. In lymphocytic-plasmacytic enterocolitis all affected samples had significant reductions in glial fibrillary acidic protein and vasoactive intestinal peptide (VIP) and mostly of neuron-specific enolase (NSE) possibly reflecting alterations in enteric glial cells and neurons. In cases with eosinophilic gastroenterocolitis significantly reduced phosphorylated neurofilament (PN) expression was present suggesting a disturbance in neuronal cytoskeleton, whereas cats with fibrosing enteropathy had reduced expression of NSE, non-phosphorylated neurofilaments (NPN), PN and VIP, possibly reflecting neuronal disturbances. In cases with intestinal lymphoma only the reduction in PN and the increase in NPN were obvious suggesting direct damage or interference of neoplastic cells with enteric neurons. In conclusion, structural and functional alterations of the ENS may contribute to clinically evident signs of vomiting and/or diarrhea.
Collapse
Affiliation(s)
- Sven Kleinschmidt
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany
| | | | | |
Collapse
|
39
|
Rodrigues DM, Li AY, Nair DG, Blennerhassett MG. Glial cell line-derived neurotrophic factor is a key neurotrophin in the postnatal enteric nervous system. Neurogastroenterol Motil 2011; 23:e44-56. [PMID: 21087354 DOI: 10.1111/j.1365-2982.2010.01626.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The enteric nervous system (ENS) continues its structural and functional growth after birth, with formation of ganglia and the innervation of growing smooth muscle. However, little is known about factors in the postnatal intestine that influence these processes. METHODS We examined the presence and potential role of glial cell line-derived nerve growth factor (GDNF) in the rat postnatal ENS using neonatal tissue, primary co-cultures of the myenteric plexus, smooth muscle, and glial cells as well as cell lines of smooth muscle or glial cells. KEY RESULTS Western blot analysis showed that GDNF and its co-receptors rearranged during transfection (RET) and GDNF family receptor alpha-1 were expressed in the muscle layer of the neonatal and adult rat intestine. Immunohistochemistry localized the receptors for GDNF to myenteric neurons, while GDNF was localized to smooth muscle cells. In a co-culture model, GDNF but not nerve growth factor, brain derived neurotrophic factor or neurotrophin-3 significantly increased neuronal survival and more than doubled the numbers of neurites in vitro. RT-PCR, qPCR, Western blotting, ELISA, and immunocytochemistry as well as bioassays of neuronal survival and of RET phosphorylation all identified intestinal smooth muscle as the source of GDNF in vitro. GDNF also induced morphological changes in the structure and organization of neurons and axons, causing marked aggregation of neuronal cell bodies and collinear development of axons. As well, GDNF (50-150 ng mL(-1)) significantly increased [(3)H]-choline uptake and stimulated [(3)H]-acetylcholine release. CONCLUSIONS & INFERENCES We conclude that GDNF derived from intestinal smooth muscle cells is a key factor influencing the structural and functional development of postnatal myenteric neurons.
Collapse
Affiliation(s)
- D M Rodrigues
- Department of Medicine, Queen's University, Kingston, ON, Canada
| | | | | | | |
Collapse
|