1
|
Saavedra LPJ, Francisco FA, Raposo SR, Cavalcante KVN, Buttow NC, Borges SC, Gomes RM, Campos HM, Gonçalves GD, Piovan S, Ghedini PC, Prates KV, Malta A, Matafome P, Mathias PCF, Almeida DL. Maternal AGE Precursors During Lactation Alters Offspring Glycemic Homeostasis Early in Life. BIOLOGY 2025; 14:160. [PMID: 40001928 PMCID: PMC11851399 DOI: 10.3390/biology14020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/29/2024] [Accepted: 01/03/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Advanced glycation end-products (AGEs) are linked to the development of oxidative stress, insulin resistance, and impaired insulin secretion. Adverse early life conditions, such as exposure to AGEs and their precursors, may lead offspring to the development of metabolic dysfunction in adulthood. Nonetheless, the early impact in offspring metabolism by maternal intake of AGEs precursors during lactation is not known. OBJECTIVE Investigate early life metabolism of the offspring whose breastfeeding dams were orally exposed to AGEs precursor. METHODS Breastfeeding Wistar rats were daily treated with the glycation precursor methylglyoxal (MG-60 mg/kg of bodyweight) by gavage or saline 0.9% control (CO) until weaning. In vivo glycemic homeostasis in male offspring was assessed, followed by euthanasia for tissue sample collection for ex vivo assessments. RESULTS At weaning, MG offspring presented decreased bodyweight (p < 0.05), perigonadal (p < 0.01) and retroperitoneal (p < 0.01) fat. MG offspring presented decreased glucose tolerance (p < 0.05), lower basal insulinemia (p < 0.001), reduced high-glucose static insulin secretion (p < 0.05), and reduced pancreatic islet area (p < 0.05). Accordingly, MG offspring pancreas showed lower GSH and SOD activity (p < 0.05; p < 0.001, respectively) and increased MPO (p < 0.05) activity. CONCLUSIONS The consumption of AGE precursors by breastfeeding dams impaired offspring pancreatic function and glycemic homeostasis early in life.
Collapse
Affiliation(s)
- Lucas P. J. Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Flávio A. Francisco
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Scarlett R. Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Keilah V. N. Cavalcante
- Department of Physiological Sciences, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (K.V.N.C.); (R.M.G.)
| | - Nilza C. Buttow
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Stephanie C. Borges
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Rodrigo M. Gomes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (K.V.N.C.); (R.M.G.)
| | - Hericles M. Campos
- Department of Pharmacology, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (H.M.C.); (P.C.G.)
| | - Gessica D. Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Paulo C. Ghedini
- Department of Pharmacology, Federal University of Goiás, Goiânia 74690-900, GO, Brazil; (H.M.C.); (P.C.G.)
| | - Kelly V. Prates
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Ananda Malta
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Paulo Matafome
- Institute of Physiology and Institute of Clinical and Biomedical Research, Faculty of Medicine and Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-447 Coimbra, Portugal;
- Coimbra Health School, ESTeSC, Instituto Politécnico de Coimbra, 3000-447 Coimbra, Portugal
- Clinical Academic Center of Coimbra, 3000-447 Coimbra, Portugal
| | - Paulo C. F. Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| | - Douglas L. Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, Maringá 87020-900, PR, Brazil; (L.P.J.S.); (F.A.F.); (S.R.R.); (N.C.B.); (S.C.B.); (G.D.G.); (S.P.); (K.V.P.); (A.M.); (P.C.F.M.)
| |
Collapse
|
2
|
Siqueira BS, Díaz Urrutia MA, Ceglarek VM, Moreira DC, Brasil Kuzniewski FT, Roberto de Souza de Almeida Leite J, Grassiolli S. A novel bombesin-related peptide modulates glucose tolerance and insulin secretion in non-obese and hypothalamic-obese rats. Toxicon 2025; 255:108230. [PMID: 39788326 DOI: 10.1016/j.toxicon.2025.108230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/10/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
This study investigated the effects of a novel bombesin-related peptide (BR-b), derived from the skin of the Chaco tree frog (Boana raniceps), on glucose homeostasis in non-obese and hypothalamic-obese male rats. Hypothalamic obesity was induced in neonatal rats through high-dose administration of monosodium glutamate (MSG; 4 g/kg), while control animals (CTL) received an equimolar saline solution. At 70 days of age, both MSG and CTL groups underwent an oral glucose tolerance test (OGTT; 2 g/kg) with or without prior intraperitoneal administration of BR-b at doses of 0.5 or 1.0 mg/kg, delivered 5 min before the glucose challenge. At 75 days of age, pancreatic islets were isolated and exposed to glucose in the presence or absence of BR-b (1.0 or 5.0 μM). MSG-treated rats developed obesity, hyperinsulinemia, and insulin resistance. BR-b administration exacerbated glucose intolerance during the OGTT, particularly at the 1.0 mg/kg dose, with more pronounced effects observed in the CTL group. Insulin secretion from pancreatic islets was influenced by both obesity status and glucose concentration. In islets from CTL rats, BR-b (5 μM) reduced insulin release under non-stimulatory glucose conditions but enhanced insulin secretion at stimulatory glucose levels. Conversely, in islets from MSG-obese rats, BR-b exhibited an inhibitory effect on insulin release at basal glucose concentrations, while the insulinotropic response to high glucose was abolished. In summary, BR-b administration shortly before the OGTT impaired glucose tolerance and modulated insulin secretion from pancreatic islets in a glucose-dependent manner in non-obese rats. These effects were attenuated or absent in MSG-obese rats, indicating that hypothalamic obesity alters the metabolic responses to bombesin-related peptides.
Collapse
Affiliation(s)
- Bruna Schumaker Siqueira
- Universidade Estadual do Oeste do Paraná, Programa de Pós-Graduação em Biociências e Saúde (PPG-BCS), Cascavel, Brazil.
| | - Marianela Andrea Díaz Urrutia
- Universidade Estadual do Oeste do Paraná, Programa de Pós-Graduação em Biociências e Saúde (PPG-BCS), Cascavel, Brazil.
| | - Vanessa Marieli Ceglarek
- Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Porto Alegre, Brazil.
| | - Daniel Carneiro Moreira
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, FM, Universidade de Brasília, Brasília, Brazil.
| | - Felipe Torres Brasil Kuzniewski
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, FM, Universidade de Brasília, Brasília, Brazil.
| | | | - Sabrina Grassiolli
- Universidade Estadual do Oeste do Paraná, Programa de Pós-Graduação em Biociências e Saúde (PPG-BCS), Cascavel, Brazil
| |
Collapse
|
3
|
Malta A, Matiusso CCI, de Lima LDS, Ribeiro TA, Tófolo LP, Almeida DL, Moreira VM, Martins IP, Pavanello A, de Freitas Mathias PC. Early exposure to a cholinergic receptor blocking agent mitigates adult obesity and protects pancreatic islet function in male rats. Am J Physiol Endocrinol Metab 2025; 328:E34-E43. [PMID: 39535172 DOI: 10.1152/ajpendo.00191.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
We tested the hypothesis that attenuation of the circulating insulin level in rats during early life can provide sustained protection against diet-induced obesity and metabolic dysfunction in adulthood. Male Wistar rats received intraperitoneal scopolamine butylbromide (SB) during the first 12 days of suckling, whereas control rats received 0.9% saline injections. The animals were weaned on day 21 and fed a normal chow diet. At 60 days of age, the control and SB groups were fed a normal chow diet (ND; 4.5% fat) or a high-fat diet (HF; 35% fat) until 90 days of age to induce obesity and metabolic dysfunction. Insulin secretion, food intake, and body weight were measured. Pancreatic islet function, autonomic nervous system function, and glucose homeostasis were evaluated at 90 days of age. During lactation, the plasma insulin concentration was significantly lower in the SB groups than in the control group. SB rats also exhibited reduced body weight. The HF diet resulted in obesity, glucose intolerance, insulin resistance, disruption of insulin secretion, and vagal hyperactivity in adult control rats. Remarkably, SB-treated rats fed the HF diet showed attenuated body weight and adiposity and did not develop diet-induced glucose/insulin imbalance. In addition, vagal activity and adequate pancreatic islet insulin secretion were preserved. Offspring exposed to SB during early life are provided with long-lasting protection against obesity and metabolic complications induced by an HF diet. An attenuated circulating insulin level in early life may have far-reaching consequences on metabolic programming.NEW & NOTEWORTHY High insulin levels during early life may lead to the late development of obesity and diabetes. We investigated whether attenuation of insulin levels by using an antimuscarinic agent could prevent diet-induced obesity and diabetes. Rats' early exposure to an antimuscarinic agent reduced insulin levels during the lactation period and promoted protection against obesity and metabolic dysfunctions. Independent of the programming mechanisms, insulin levels during early life may be a defining factor of health or diseases later in life.
Collapse
Affiliation(s)
- Ananda Malta
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | | | - Lucas da Silva de Lima
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Tatiane Aparecida Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Department of Biochemistry and Biomedical Science, McMaster University, Hamilton, Ontario, Canada
| | - Laize Peron Tófolo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Physical Education Department, Paraná State University, Paranavai, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
| | - Isabela Peixoto Martins
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Health Sciences Center, Unicesumar, Maringa, Brazil
| | - Audrei Pavanello
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringa, Maringa, Brazil
- Health Sciences Center, Unicesumar, Maringa, Brazil
| | | |
Collapse
|
4
|
de Freitas Mathias PC, Dantas Rodrigues AM, Lisboa PC, Miranda RA, Malta A, Ribeiro TA, Barella LF, Dias G, Lima TAL, Gomes RM, de Moura EG, de Oliveira JC. Maternal Low-Protein Diet During Nursing Leads to Glucose-Insulin Dyshomeostasis and Pancreatic-Islet Dysfunction by Disrupting Glucocorticoid Responsiveness in Male Rats. BIOLOGY 2024; 13:1036. [PMID: 39765703 PMCID: PMC11673749 DOI: 10.3390/biology13121036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025]
Abstract
Both perinatal malnutrition and elevated glucocorticoids are pivotal triggers of the growing global pandemic of metabolic diseases. Here, we studied the effects of metabolic stress responsiveness on glucose-insulin homeostasis and pancreatic-islet function in male Wistar offspring whose mothers underwent protein restriction during lactation. During the first two weeks after delivery, lactating dams were fed a low-protein (4% protein, LP group) or normal-protein diet (22.5% protein, NP group). At 90 days of age, male rat offspring were challenged with food deprivation (72 h of fasting), intracerebroventricular (icv) injection of dexamethasone (2 µL, 2.115 mmol/L) or chronic intraperitoneal injection of dexamethasone (1 mg/kg body weight/5 days). Body weight, food intake, intravenous glucose tolerance test (ivGTT) results, insulin secretion and biochemical parameters were assessed. LP rats did not display significant metabolic changes after long-term starvation (p > 0.05) or under the central effect of dexamethasone (p = 0.999). Chronic dexamethasone induced rapid hyperglycemia (~1.2-fold, p < 0.001) and hyperinsulinemia (NP: 65%; LP: 216%; p < 0.001), decreased insulin sensitivity (NP: ~2-fold; LP: ~4-fold; p < 0.001), reduced insulinemia (20%) and increased glycemia (35%) only in NP rats under ivGTT conditions (p < 0.001). Glucose and acetylcholine insulinotropic effects, as well as the muscarinic receptor antagonist response, were reduced by chronic dexamethasone only in pancreatic islets from NP rats (p < 0.05). The direct effect of dexamethasone on pancreatic islets reduced insulin secretion (NP: 60.2%, p < 0.001; LP: 33.8%, p < 0.001). Peripheral glucose-insulin dyshomeostasis and functional failure of pancreatic islets in LP rats, as evidenced by an impaired acute and chronic response to metabolic stress, may be due to excessive corticosterone action as a long-term consequence.
Collapse
Affiliation(s)
- Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá 87020-900, Brazil; (P.C.d.F.M.); (A.M.); (T.A.R.); (L.F.B.)
| | - Aline Milena Dantas Rodrigues
- Research Group on Perinatal Programming of Metabolic Diseases: DOHaD Paradigm, Laboratory of Metabolic and Cardiovascular Diseases, Health Education and Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, University Campus of Sinop, Sinop 78556-264, Brazil; (A.M.D.R.); (G.D.); (T.A.L.L.)
| | - Patrícia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (P.C.L.); (R.A.M.); (E.G.d.M.)
| | - Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (P.C.L.); (R.A.M.); (E.G.d.M.)
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá 87020-900, Brazil; (P.C.d.F.M.); (A.M.); (T.A.R.); (L.F.B.)
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá 87020-900, Brazil; (P.C.d.F.M.); (A.M.); (T.A.R.); (L.F.B.)
| | - Luiz Felipe Barella
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá 87020-900, Brazil; (P.C.d.F.M.); (A.M.); (T.A.R.); (L.F.B.)
| | - Ginislene Dias
- Research Group on Perinatal Programming of Metabolic Diseases: DOHaD Paradigm, Laboratory of Metabolic and Cardiovascular Diseases, Health Education and Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, University Campus of Sinop, Sinop 78556-264, Brazil; (A.M.D.R.); (G.D.); (T.A.L.L.)
| | - Thalyne Aparecida Leite Lima
- Research Group on Perinatal Programming of Metabolic Diseases: DOHaD Paradigm, Laboratory of Metabolic and Cardiovascular Diseases, Health Education and Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, University Campus of Sinop, Sinop 78556-264, Brazil; (A.M.D.R.); (G.D.); (T.A.L.L.)
| | - Rodrigo Mello Gomes
- Laboratory of Endocrine Physiology and Metabolism, Institute of Biological Sciences, Federal University of Goiás, Goiânia 74690-900, Brazil;
| | - Egberto Gaspar de Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, State University of Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (P.C.L.); (R.A.M.); (E.G.d.M.)
| | - Júlio Cezar de Oliveira
- Research Group on Perinatal Programming of Metabolic Diseases: DOHaD Paradigm, Laboratory of Metabolic and Cardiovascular Diseases, Health Education and Research Center (NUPADS), Institute of Health Sciences, Federal University of Mato Grosso, University Campus of Sinop, Sinop 78556-264, Brazil; (A.M.D.R.); (G.D.); (T.A.L.L.)
| |
Collapse
|
5
|
Fernandes‐da‐Silva A, Miranda RA, Lisboa PC, Souza‐Mello V. Revisiting pancreatic islet isolation in murine models: A practical and effective technical protocol. Physiol Rep 2024; 12:e16040. [PMID: 38725080 PMCID: PMC11082087 DOI: 10.14814/phy2.16040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/12/2024] Open
Abstract
The endocrine pancreas is composed of clusters of cell groups called pancreatic islets. These cells are responsible for the synthesis and secretion of hormones crucial for glycemic homeostasis, such as insulin and glucagon. Therefore, these cells were the targets of many studies. One method to study and/or understand endocrine pancreatic physiology is the isolation of these islets and stimulation of hormone production using different concentrations of glucose, agonists, and/or antagonists of specific secretagogues and mimicking the stimulation of hormonal synthesis and secretion. Many researchers studied pancreatic physiology in murine models due to their ease of maintenance and rapid development. However, the isolation of pancreatic islets involves meticulous processes that may vary between rodent species. The present study describes a simple and effective technical protocol for isolating intact islets from mice and rats for use as a practical guide for researchers. The method involves digestion of the acinar parenchyma by intraductal collagenase. Isolated islets are suitable for in vitro endocrine secretion analyses, microscopy techniques, and biochemical analyses.
Collapse
Affiliation(s)
- Aline Fernandes‐da‐Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Patricia Cristina Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| | - Vanessa Souza‐Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology Roberto Alcantara GomesState University of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
6
|
Barssotti L, Soares GM, Marconato-Júnior E, Lourençoni Alves B, Oliveira KM, Carneiro EM, Boschero AC, Barbosa HCL. KSRP improves pancreatic beta cell function and survival. Sci Rep 2024; 14:6136. [PMID: 38480757 PMCID: PMC10937633 DOI: 10.1038/s41598-024-55505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/24/2024] [Indexed: 03/17/2024] Open
Abstract
Impaired insulin production and/or secretion by pancreatic beta cells can lead to high blood glucose levels and type 2 diabetes (T2D). Therefore, investigating new proteins involved in beta cell response to stress conditions could be useful in finding new targets for therapeutic approaches. KH-type splicing regulatory protein (KSRP) is a protein usually involved in gene expression due to its role in post-transcriptional regulation. Although there are studies describing the important role of KSRP in tissues closely related to glucose homeostasis, its effect on pancreatic beta cells has not been explored so far. Pancreatic islets from diet-induced obese mice (C57BL/6JUnib) were used to determine KSRP expression and we also performed in vitro experiments exposing INS-1E cells (pancreatic beta cell line) to different stressors (palmitate or cyclopiazonic acid-CPA) to induce cellular dysfunction. Here we show that KSRP expression is reduced in all the beta cell dysfunction models tested. In addition, when manipulated to knock down KSRP, beta cells exhibited increased death and impaired insulin secretion, whereas KSRP overexpression prevented cell death and increased insulin secretion. Taken together, our findings suggest that KSRP could be an important target to protect beta cells from impaired functioning and death.
Collapse
Affiliation(s)
- Leticia Barssotti
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Gabriela Moreira Soares
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Emílio Marconato-Júnior
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Bruna Lourençoni Alves
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Kênia Moreno Oliveira
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Everardo Magalhães Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Antonio Carlos Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil
| | - Helena Cristina Lima Barbosa
- Obesity and Comorbidities Research Center (OCRC), Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, 13083864, Brazil.
| |
Collapse
|
7
|
de Jesus DS, Bargi-Souza P, Cruzat V, Yechoor V, Carpinelli AR, Peliciari-Garcia RA. BMAL1 modulates ROS generation and insulin secretion in pancreatic β-cells: An effect possibly mediated via NOX2. Mol Cell Endocrinol 2022; 555:111725. [PMID: 35868425 DOI: 10.1016/j.mce.2022.111725] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022]
Abstract
The pancreatic β cells circadian clock plays a relevant role in glucose metabolism. NADPH oxidase (NOX) family is responsible for producing reactive oxygen species (ROS), such as superoxide anion and hydrogen peroxide, using NADPH as an electron donor. In pancreatic β-cells, NOX-derived ROS inhibits basal and glucose-stimulated insulin secretion. Thus, we hypothesized that the absence of BMAL1, a core circadian clock component, could trigger an increase of NOX2-derived ROS in pancreatic β cells, inhibiting insulin secretion under basal and stimulated glucose conditions. To test such hypothesis, Bmal1 knockdown (KD) was performed in cultured clonal β-cell line (INS-1E) and knocked out in isolated pancreatic islets, using a tissue-specific β-cells Bmal1 knockout (KO) mice. The insulin secretion was assessed in the presence of NOX inhibitors. The Bmal1 KD within INS-1E cells elicited a rise of intracellular ROS content under both glucose stimuli (2.8 mM and 16.7 mM), associated with an increase in Nox2 expression. Additionally, alterations of glutathione levels, CuZnSOD and catalase activities, reduction of ATP/ADP ratio, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and aconitase activities, followed by glucokinase and Slc2a2 (Glut2) expression were also observed in INS-1E β-cells, reflecting in a diminished insulin secretion pattern. The isolated islets from β-cell Bmal1-/- mice have shown a similar cellular response, where an increased NOX2-derived ROS content and a reduced basal- and glucose-stimulated insulin secretion were observed. Therefore, together with NOX inhibition (Apocynin), polyethene-glycol linked to superoxide dismutase (PEG-SOD), phorbol myristate acetate (PMA), and diethyldithiocarbamate (DDC) data, our findings suggest a possible BMAL1-mediated NOX2-derived ROS generation in pancreatic β cells, leading to the modulation of both basal- and glucose-stimulated insulin secretion.
Collapse
Affiliation(s)
- Daniel Simoes de Jesus
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo (USP), SP, Brazil; Centre for Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Paula Bargi-Souza
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Vinicius Cruzat
- Faculty of Health, Torrens University, Melbourne, Victoria, Australia
| | - Vijay Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo (USP), SP, Brazil
| | - Rodrigo Antonio Peliciari-Garcia
- Department of Biological Sciences, Morphophysiology and Pathology Sector, Federal University of São Paulo (UNIFESP), Diadema, SP, Brazil.
| |
Collapse
|
8
|
Prates KV, Pavanello A, Gongora AB, Moreira VM, de Moraes AMP, Rigo KP, Vieira E, Mathias PCDF. Time-restricted feeding during embryonic development leads to metabolic dysfunction in adult rat offspring. Nutrition 2022; 103-104:111776. [DOI: 10.1016/j.nut.2022.111776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/24/2022]
|
9
|
Valle MMR, Vilas-Boas EA, Lucena CF, Teixeira SA, Muscara MN, Carpinelli AR. Metformin disrupts insulin secretion, causes proapoptotic and oxidative effects in rat pancreatic beta-cells in vitro. J Biochem Mol Toxicol 2022; 36:e23007. [PMID: 35199402 DOI: 10.1002/jbt.23007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 11/28/2021] [Accepted: 01/04/2022] [Indexed: 11/07/2022]
Abstract
Metformin is the first-line drug to treat type 2 diabetes mellitus. Its mechanism of action is still debatable, and recent studies report that metformin attenuates oxidative stress. This study evaluated the in vitro antioxidant effects of a broad range of metformin concentrations on insulin-producing cells. The cell cycle, metabolism, glucose-stimulated insulin secretion, and cell death were evaluated to determine the biguanide effects on beta-cell function and survival. Antioxidant potential was based on reactive oxygen species (ROS), reduced glutathione (GSH), oxidative stress biomarker levels, and antioxidant enzyme and transcriptional factor Nrf2 activities. The results demonstrate that metformin disrupted GSIS in a concentration-dependent manner, lowered insulin content, and attenuated beta-cell metabolism. At high concentrations, metformin induced cell death and cell cycle arrest as well as increased ROS generation, consequently reducing GSH content. Although carbonylated protein content was elevated, indicating oxidative stress, the antioxidant enzyme and Nrf2 activities were not altered. In conclusion, our results show that metformin disrupts pancreatic beta-cell functionality but does not exert a putative antioxidant effect. It is important to note that the drug could potentially affect beta-cells, especially at high circulating levels.
Collapse
Affiliation(s)
- Maíra M R Valle
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Camila F Lucena
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| | - Simone A Teixeira
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil.,Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcelo N Muscara
- Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil.,Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Angelo R Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Physiology, Division of Biological Sciences, Federal University of Parana, Curitiba, Parana, Brazil
| |
Collapse
|
10
|
Veras K, Lucena CF, Goedcke J, Evangelista FS, Carpinelli A, Carvalho CRDO. Moderate Exercise Training Combined With a High-Fat and Sucrose Diet Protects Pancreatic Islet Function in Male C57BL/6J Mice. Front Endocrinol (Lausanne) 2022; 13:881236. [PMID: 35669687 PMCID: PMC9165053 DOI: 10.3389/fendo.2022.881236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity is mainly caused by excess energy intake and physical inactivity, and the number of overweight/obese individuals has been steadily increasing for decades. Previous studies showed that rodents fed westernized diets exhibit endocrine pancreas deterioration and a range of metabolic disorders. This study evaluated the effects of moderated aerobic treadmill exercise training on pancreatic islet cell viability and function in mice consuming a high-fat and sucrose diet. In the present study, 60-day-old male C57BL/6J mice were divided into four groups: control (C), fed a standard diet AIN-93M (3.83 kcal/g; 70% carbohydrate (cornstarch and dextrinized starch were chosen as the major source of carbohydrate for the AIN-93 diet. In addition, a small amount of sucrose), 20% protein (casein), and 10% fat (soybean) with no training (i.e., sedentary); C + training (CTR, fed the standard diet with eight weeks of exercise; high-fat diet + sucrose (HFDS), fed a high fat and sucrose diet (5.2 kcal/g; 20% carbohydrate (cornstarch and dextrinized starch were chosen as the major source of carbohydrate), 20% protein (casein), 60% fat (Lard was chosen as the major source of fat and a small amount of soybean) + 20% sucrose diluted in drinking water with no training; and HFDS + training (HFDSTR). After eight weeks, the HFDS mice displayed increased body weight (P<0.001) and epididymal, inguinal and retroperitoneal adipose tissue mass (P<0.01). These mice also presented insulin resistance (P<0.01), glucose intolerance (P<0.001), impaired glucose-stimulated insulin secretion (GSIS) and were less responsive to the physiological net ROS production induced by glucose stimulus. The HFDS group's pancreatic islet cells were 38% less viable and 59% more apoptotic than those from the C group (P<0.05). The HFDSTR improved glucose tolerance, body mass, insulin sensitivity and GSIS (P<0.05). Furthermore, HFDSTR mice had 53% more viable isolated pancreatic islets cells and 29% fewer apoptotic cells than the HFDS group (P<0.01). Thus, exercise training may slow down and/or prevent adverse metabolic effects associated with consuming a westernized diet.
Collapse
Affiliation(s)
- Katherine Veras
- Institute of Biomedical Sciences, Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | - Camila Ferraz Lucena
- Institute of Biomedical Sciences, Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | - Julia Goedcke
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | | | - Angelo Carpinelli
- Institute of Biomedical Sciences, Department of Physiology and Biophysics, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
11
|
Freitas-Dias R, Lima TI, Costa-Junior JM, Gonçalves LM, Araujo HN, Paula FMM, Santos GJ, Branco RCS, Ou K, Kaestner KH, Silveira LR, Oliveira CAM, Boschero AC, Zoppi CC, Carneiro EM. Offspring from trained male mice inherit improved muscle mitochondrial function through PPAR co-repressor modulation. Life Sci 2021; 291:120239. [PMID: 34942163 DOI: 10.1016/j.lfs.2021.120239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/03/2021] [Accepted: 12/11/2021] [Indexed: 10/19/2022]
Abstract
Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.
Collapse
Affiliation(s)
- Ricardo Freitas-Dias
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physical Therapy, Laboratory of Exercise Physiology, University of Pernambuco, Petrolina, PE, Brazil
| | - Tanes I Lima
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jose Maria Costa-Junior
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Luciana M Gonçalves
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Hygor N Araujo
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Flavia M M Paula
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gustavo J Santos
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina (UFSC), Florianopolis, SC, Brazil
| | - Renato Chaves Souto Branco
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kristy Ou
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Leonardo R Silveira
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Camila A M Oliveira
- Department of Biosciences, Federal University of Sao Paulo, Santos, SP, Brazil
| | - Antonio C Boschero
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Claudio C Zoppi
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| | - Everardo M Carneiro
- Obesity and Comorbidities Research Center (OCRC), Department of Functional and Structural Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
12
|
Villaça CDBP, de Paula CC, de Oliveira CC, Vilas-Boas EA, Dos Santos-Silva JC, de Oliveira SF, Abdulkader F, Ferreira SM, Ortis F. Beneficial effects of physical exercise for β-cell maintenance in a type 1 diabetes mellitus animal model. Exp Physiol 2021; 106:1482-1497. [PMID: 33913203 DOI: 10.1113/ep088872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 04/16/2021] [Indexed: 12/17/2022]
Abstract
NEW FINDINGS What is the central question of this study? Type 1 diabetes mellitus (T1D) leads to hyperglycaemia owing to pancreatic β-cell destruction by the immune system. Physical exercise has been shown to have potentially beneficial protective roles against cytokine-induced pancreatic β-cell death, but its benefits are yet to be proved and should be understood better, especially in the islet environment. What is the main finding and its importance? Physical exercise protects against β-cell loss in a well-described animal model for T1D, induced by multiple low doses of streptozotocin. This seems to be related to reduced cytokine-induced β-cell death and increased islet cell proliferation. Contributions of islet neogenesis and/or transdifferentiation of pancreatic non-β-cells into β-cells cannot be excluded. ABSTRACT Physical exercise has beneficial effects on pancreatic β-cell function and survival in a pro-inflammatory environment. Although these effects have been linked to decreased islet inflammation and modulation of pro-apoptotic pathways, little is known about the islet microenvironment. Our aim was to evaluate the effects of physical exercise in islet histomorphology in a mouse model of type 1 diabetes mellitus induced by multiple low doses of streptozotocin. As expected, induction of type 1 diabetes mellitus led to β-cell loss and, consequently, decreased islet area. Interestingly, although the decrease in islet area was not prevented by physical exercise, this was not the case for the decrease in β-cell mass. This was probably related to induction of β-cell regeneration, because we observed increased proliferation and regeneration markers, such as Ki67 and Pcna, in islets of trained mice. These were found in the central and peripheral regions of the islets. An increase in the percentage of α- and δ-cells in these conditions, combined with an increase in proliferation and Pax4 labelling in peripheral regions, suggest that β-cell regeneration might also occur by transdifferentiation. This agrees with the presence of cells double stained for insulin and glucagon only in islets of diabetic trained mice. In addition, this group had more extra-islet insulin-positive cells and islets associated with ducts than diabetic mice. Physical exercise also decreased nuclear factor-κB activation in islet cells of diabetic trained compared with diabetic untrained mice, indicating a decrease in pro-inflammatory cytokine-induced β-cell death. Taken together, these findings indicate that preservation of β-cell mass induced by physical exercise involves an increase in β-cell replication and decrease in β-cell death, together with islet neogenesis and islet cell transdifferentiation.
Collapse
Affiliation(s)
| | - Carolina Cavalcante de Paula
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| | - Caroline Cruz de Oliveira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| | | | - Sérgio Ferreira de Oliveira
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| | - Fernando Abdulkader
- Department of Physiology and Biophysics, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| | - Sandra Mara Ferreira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, Brazil
| | - Fernanda Ortis
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences (ICB), University of São Paulo (USP), São Paulo, Brazil
| |
Collapse
|
13
|
Amino acid restriction alters survival mechanisms in pancreatic beta cells: possible role of the PI3K/Akt pathway. Eur J Nutr 2021; 60:3947-3957. [PMID: 33913012 PMCID: PMC8081284 DOI: 10.1007/s00394-021-02568-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 04/16/2021] [Indexed: 11/25/2022]
Abstract
Background and aims Malnutrition in the early stages of life may lead to changes in the glycemic metabolism during adulthood, such as pancreatic beta cells dysfunction and failure. Therefore, this study aimed to evaluate the effects of an in vitro amino acid restriction model on the function and viability of pancreatic beta cells. Methods Insulin-producing cells (INS-1E) were maintained in control or amino acid restricted culture medium containing 1 × or 0.25 × of amino acids, respectively, for 48 h. Results Amino acid restricted group showed lower insulin secretion and insulin gene expression, reduced mitochondrial oxygen consumption rate and reactive oxygen species production. Besides, amino acid restricted group also showed higher levels of endoplasmic reticulum stress and apoptosis markers and enhanced Akt phosphorylation. However, even with higher levels of apoptosis markers, amino acid restricted group did not show higher levels of cell death unless the PI3K/Akt pathway was inhibited. Conclusion Amino acid restricted beta cell viability seems to be dependent on the PI3K/Akt pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s00394-021-02568-2.
Collapse
|
14
|
Resistance exercise training improves glucose homeostasis by enhancing insulin secretion in C57BL/6 mice. Sci Rep 2021; 11:8574. [PMID: 33883630 PMCID: PMC8060292 DOI: 10.1038/s41598-021-88105-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/06/2021] [Indexed: 01/14/2023] Open
Abstract
Resistance exercise exerts beneficial effects on glycemic control, which could be mediated by exercise-induced humoral factors released in the bloodstream. Here, we used C57Bl/6 healthy mice, submitted to resistance exercise training for 10 weeks. Trained mice presented higher muscle weight and maximum voluntary carrying capacity, combined with reduced body weight gain and fat deposition. Resistance training improved glucose tolerance and reduced glycemia, with no alterations in insulin sensitivity. In addition, trained mice displayed higher insulinemia in fed state, associated with increased glucose-stimulated insulin secretion. Islets from trained mice showed reduced expression of genes related to endoplasmic reticulum (ER) stress, associated with increased expression of Ins2. INS-1E beta-cells incubated with serum from trained mice displayed similar pattern of insulin secretion and gene expression than isolated islets from trained mice. When exposed to CPA (an ER stress inducer), the serum from trained mice partially preserved the secretory function of INS-1E cells, and prevented CPA-induced apoptosis. These data suggest that resistance training, in healthy mice, improves glucose homeostasis by enhancing insulin secretion, which could be driven, at least in part, by humoral factors.
Collapse
|
15
|
Milani PG, Piovan S, Lima YC, Zorzenon MRT, da Rosa CVD, Peixoto GML, de Freitas Mathias PC, Natali MRM, da Costa SC, Mareze-Costa CE. Whey protein enriched with Stevia rebaudiana fraction restores the pancreatic function of streptozotocin induced diabetic rats. Journal of Food Science and Technology 2021; 58:805-810. [PMID: 33568874 DOI: 10.1007/s13197-020-04799-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023]
Abstract
The study aims to analyse the treatment of whey protein enriched with Stevia rebaudiana fraction in insulin secretion and its role mitigating streptozotocin-induced hyperglycemia in rats. Thus, diabetic animals were treated with whey protein enriched with S. rebaudiana fraction or with only the protein isolate or only the Stevia fraction. Insulin level in plasma was measured by radioimmunoassay and the viability of β cells was detected by immunohistochemistry. The results showed that diabetic animals treated with whey protein enriched with S. rebaudiana fraction had a greater recovery from insulinemia, with plasma levels similar to non-diabetic animals (~ 0.13 ng/mL). In addition, the same group showed a higher number of insulin-positive pancreatic B cells (~ 66%) in immunohistochemistry analysis, while the diabetic groups treated with only the fraction of stevia or whey protein showed 38 and 59% of positive cells, respectively. These results show that the treatment may have restored the viability of streptozotocin-injured pancreatic B cells, and consequently increased insulin secretion, suggesting whey protein enriched with S. rebaudiana fraction can be used an adjunct/supplement in diabetic treatment.
Collapse
Affiliation(s)
| | - Silvano Piovan
- Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR Brazil
| | - Yago Carvalho Lima
- Post-Graduate Program in Sciences (Human Physiology), State University of Sao Paulo, São Paulo, SP Brazil
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Pereira de Arruda EH, Vieira da Silva GL, da Rosa-Santos CA, Arantes VC, de Barros Reis MA, Colodel EM, Gaspar de Moura E, Lisboa PC, Carneiro EM, Damazo AS, Latorraca MQ. Protein restriction during pregnancy impairs intra-islet GLP-1 and the expansion of β-cell mass. Mol Cell Endocrinol 2020; 518:110977. [PMID: 32791189 DOI: 10.1016/j.mce.2020.110977] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 12/26/2022]
Abstract
We evaluated whether protein restriction during pregnancy alters the morphometry of pancreatic islets, the intra-islet glucagon-like peptide-1 (GLP-1) production, and the anti-apoptotic signalling pathway modulated by GLP-1. Control non-pregnant (CNP) and control pregnant (CP) rats were fed a 17% protein diet, and low-protein non-pregnant (LPNP) and low-protein pregnant (LPP) groups were fed a 6% protein diet. The masses of islets and β-cells were similar in the LPNP group and the CNP group but were higher in the CP group than in the CNP group and were equal in the LPP group and the LPNP group. Both variables were lower in the LPP group than in the CP group. Prohormone convertase 2 and GLP-1 fluorescence in α-cells was lower in the low-protein groups than in the control groups. The least PC2/glucagon colocalization was observed in the LPP group, and the most was observed in the CP group. There was less prohormone convertase 1/3/glucagon colocalization in the LPP group than in the CP group. GLP-1/glucagon colocalization was similar in the LPP, CP and CNP groups, which showed less GLP-1/glucagon colocalization than the LPNP group. The mRNA Pka, Creb and Pdx-1 contents were higher in islets from pregnant rats than in islets from non-pregnant rats. Protein restriction during pregnancy impaired the mass of β-cells and the intra-islet GLP-1 production but did not interfere with the transcription of genes of the anti-apoptotic signalling pathway modulated by GLP-1.
Collapse
Affiliation(s)
| | | | - Chaiane Aline da Rosa-Santos
- Mestrado em Nutrição, Alimentos e Metabolismo, Faculdade de Nutrição, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | - Vanessa Cristina Arantes
- Departamento de Alimentos e Nutrição, Faculdade de Nutrição, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | | | - Edson Moleta Colodel
- Departamento de Clínica Médica Veterinária, Faculdade de Agronomia e Medicina Veterinária, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | - Egberto Gaspar de Moura
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patrícia Cristina Lisboa
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Everardo Magalhães Carneiro
- Departamento de Anatomia, Biologia Cellular, Fisiologia e Biofísica, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, SP, Brazil
| | - Amílcar Sabino Damazo
- Departamento de Ciências Básicas da Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | - Márcia Queiroz Latorraca
- Departamento de Alimentos e Nutrição, Faculdade de Nutrição, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil.
| |
Collapse
|
17
|
Protein malnutrition early in life increased apoptosis but did not alter the β-cell mass during gestation. Br J Nutr 2020; 125:1111-1124. [PMID: 32912341 DOI: 10.1017/s0007114520003554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
We evaluated whether early-life protein restriction alters structural parameters that affect β-cell mass on the 15th day and 20th day of gestation in control pregnant (CP), control non-pregnant (CNP), low-protein pregnant (LPP) and low-protein non-pregnant (LPNP) rats from the fetal to the adult life stage as well as in protein-restricted rats that recovered after weaning (recovered pregnant (RP) and recovered non-pregnant). On the 15th day of gestation, the CNP group had a higher proportion of smaller islets, whereas the CP group exhibited a higher proportion of islets larger than the median. The β-cell mass was lower in the low-protein group than that in the recovered and control groups. Gestation increased the β-cell mass, β-cell proliferation frequency and neogenesis frequency independently of the nutritional status. The apoptosis frequency was increased in the recovered groups compared with that in the other groups. On the 20th day of gestation, a higher proportion of islets smaller than the median was observed in the non-pregnant groups, whereas a higher proportion of islets larger than the median was observed in the RP, LPP and CP groups. β-Cell mass was lower in the low-protein group than that in the recovered and control groups, regardless of the physiological status. The β-cell proliferation frequency was lower, whereas the apoptosis rate was higher in recovered rats compared with those in the low-protein and control rats. Thus, protein malnutrition early in life did not alter the mass of β-cells, especially in the first two-thirds of gestation, despite the increase in apoptosis.
Collapse
|
18
|
Panveloski-Costa AC, Kuwabara WMT, Munhoz AC, Lucena CF, Curi R, Carpinelli AR, Nunes MT. The insulin resistance is reversed by exogenous 3,5,3'triiodothyronine in type 2 diabetic Goto-Kakizaki rats by an inflammatory-independent pathway. Endocrine 2020; 68:287-295. [PMID: 31997150 DOI: 10.1007/s12020-020-02208-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/17/2020] [Indexed: 11/30/2022]
Abstract
PURPOSE Diabetes mellitus (DM) has a multifactorial etiology that imparts a particular challenge to effective pharmacotherapy. Thyroid hormone actions have demonstrated beneficial effects in diabetic as well as obese rats. In both conditions, inflammation status plays a crucial role in the development of insulin resistance. Taking this into consideration, the present study aimed to demonstrate another possible pathway of thyroid hormone action on insulin sensitivity in a spontaneous type 2 diabetic rat model: the Goto-Kakizaki (GK) rats. GK animals present all typical hallmarks of type 2 DM (T2DM), except the usual peripheric inflammatory condition, observed in the other T2DM animal models. METHODS GK rats were treated or not with 3,5,3'triiodothyronine (T3). Insulin sensitivity, glucose tolerance, and proteins related to glucose uptake and utilization were evaluated in the skeletal muscle, white adipose tissue, and liver. RESULTS GK rats T3-treated presented enhanced insulin sensitivity, increased GLUT-4 content in the white adipose tissue and skeletal muscle, and increased hexokinase and citrate synthase content in skeletal muscle. Both non-treated and T3-treated GK rats did not present alterations in cytokine content in white adipose tissue, skeletal muscle, liver, and serum. CONCLUSIONS These results indicate that T3 improves insulin sensitivity in diabetic rats by a novel inflammatory-independent mechanism.
Collapse
Affiliation(s)
- Ana Carolina Panveloski-Costa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | - Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila Ferraz Lucena
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rui Curi
- Interdisciplinar Health Science Post-Graduate Program, Cruzeiro do Sul University, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Tereza Nunes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Munhoz AC, Vilas-Boas EA, Panveloski-Costa AC, Leite JSM, Lucena CF, Riva P, Emilio H, Carpinelli AR. Intermittent Fasting for Twelve Weeks Leads to Increases in Fat Mass and Hyperinsulinemia in Young Female Wistar Rats. Nutrients 2020; 12:E1029. [PMID: 32283715 PMCID: PMC7230500 DOI: 10.3390/nu12041029] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023] Open
Abstract
Fasting is known to cause physiological changes in the endocrine pancreas, including decreased insulin secretion and increased reactive oxygen species (ROS) production. However, there is no consensus about the long-term effects of intermittent fasting (IF), which can involve up to 24 hours of fasting interspersed with normal feeding days. In the present study, we analyzed the effects of alternate-day IF for 12 weeks in a developing and healthy organism. Female 30-day-old Wistar rats were randomly divided into two groups: control, with free access to standard rodent chow; and IF, subjected to 24-hour fasts intercalated with 24-hours of free access to the same chow. Alternate-day IF decreased weight gain and food intake. Surprisingly, IF also elevated plasma insulin concentrations, both at baseline and after glucose administration collected during oGTT. After 12 weeks of dietary intervention, pancreatic islets displayed increased ROS production and apoptosis. Despite their lower body weight, IF animals had increased fat reserves and decreased muscle mass. Taken together, these findings suggest that alternate-day IF promote β -cell dysfunction, especially in developing animals. More long-term research is necessary to define the best IF protocol to reduce side effects.
Collapse
Affiliation(s)
- Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Eloisa Aparecida Vilas-Boas
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Ana Carolina Panveloski-Costa
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Jaqueline Santos Moreira Leite
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Camila Ferraz Lucena
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Patrícia Riva
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| | - Henriette Emilio
- Department of General Biology, Ponta Grossa State University, 4748 General Carlos Cavalcanti avenue, Uvaranas, Parana, PR 84030-900, Brazil;
| | - Angelo R. Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, 1524 Professor Lineu Prestes avenue, Butanta, São Paulo 05508-900, Brazil; (E.A.V.-B.); (A.C.P.-C.); (J.S.M.L.); (C.F.L.); (P.R.); (A.R.C.)
| |
Collapse
|
20
|
Cholinergic-pathway-weakness-associated pancreatic islet dysfunction: a low-protein-diet imprint effect on weaned rat offspring. J Dev Orig Health Dis 2020; 11:484-491. [DOI: 10.1017/s2040174420000215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AbstractCurrently, metabolic disorders are one of the major health problems worldwide, which have been shown to be related to perinatal nutritional insults, and the autonomic nervous system and endocrine pancreas are pivotal targets of the malprogramming of metabolic function. We aimed to assess glucose–insulin homeostasis and the involvement of cholinergic responsiveness (vagus nerve activity and insulinotropic muscarinic response) in pancreatic islet capacity to secrete insulin in weaned rat offspring whose mothers were undernourished in the first 2 weeks of the suckling phase. At delivery, dams were fed a low-protein (4% protein, LP group) or a normal-protein diet (20.5% protein, NP group) during the first 2 weeks of the suckling period. Litter size was adjusted to six pups per mother, and rats were weaned at 21 days old. Weaned LP rats presented a lean phenotype (P < 0.01); hypoglycaemia, hypoinsulinaemia and hypoleptinaemia (P < 0.05); and normal corticosteronaemia (P > 0.05). In addition, milk insulin levels in mothers of the LP rats were twofold higher than those of mothers of the NP rats (P < 0.001). Regarding glucose–insulin homeostasis, weaned LP rats were glucose-intolerant (P < 0.01) and displayed impaired pancreatic islet insulinotropic function (P < 0.05). The M3 subtype of the muscarinic acetylcholine receptor (M3mAChR) from weaned LP rats was less responsive, and the superior vagus nerve electrical activity was reduced by 30% (P < 0.01). A low-protein diet in the suckling period malprogrammes the vagus nerve to low tonus and impairs muscarinic response in the pancreatic β-cells of weaned rats, which are imprinted to secrete inadequate insulin amounts from an early age.
Collapse
|
21
|
Valério Prates K, Ribeiro TA, Pavanello A, Jacinto Saavedra LP, Moreira VM, da Silva Silveira S, Martins IP, Francisco FA, Ferreira Junior MD, Alves VS, Tófolo LP, Previate C, da Silva Franco CC, Gomes RM, Palma-Rigo K, Malta A, de Freitas Mathias PC. Potential attenuation of early-life overfeeding-induced metabolic dysfunction by chronic maternal acetylcholinesterase inhibitor exposure. Toxicology 2019; 425:152250. [PMID: 31326399 DOI: 10.1016/j.tox.2019.152250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 12/12/2022]
Abstract
Evidence suggests that low concentration perinatal exposure to environmental contaminants, such as organophosphate (OP) is associated with later life insulin resistance and type 2 diabetes. The aim of this work was to investigate whether chronic maternal OP exposure exacerbates metabolic dysfunctions in early-overfed rats. During pregnancy and lactational periods, dams received OP by gavage. To induce neonatal overnutrition at postnatal day 3, pups were standardized to 9 or 3 per nest. At 90-days-old, glucose-insulin homeostasis and insulin release from pancreatic islets were analyzed. While both OP exposure and overfeeding alone did induce diabetogenic phenotypes in adulthood, there was no exacerbation in rats that experienced both. Unexpectedly, the group that experienced both had improved adiposity, metabolic parameters, attenuated insulin release from isolated islets in the presence of glucose and low function of muscarinic acetylcholine receptor M3, as well as an attenuation of beta cell mass hyperplasia. High levels of butyrylcholinesterase and low levels of insulin in milk may contribute to the OP-induced developmental programming. Our study showed that maternal OP exposure may program insulin release as well as endocrine pancreas structure, thus affecting metabolism in adulthood. Our data suggest that while perinatal OP exposure alone increases the risk for later life T2D, it actually reverses many of the programmed metabolic dysfunction that is induced by postnatal overfeeding. These surprising results may suggest that low-dose administration of acetylcholinesterase inhibitors could be of utility in preventing detrimental developmental programming that is caused by early-life overnutrition.
Collapse
Affiliation(s)
- Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil.
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Sandra da Silva Silveira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | | | - Vander Silva Alves
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Carina Previate
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Federal University of Goias, Goiania, GO, Brazil
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringa, Parana, Brazil
| |
Collapse
|
22
|
Moreira VM, Almeida D, da Silva Franco CC, Gomes RM, Palma-Rigo K, Prates KV, Tófolo LP, Malta A, Francisco FA, Pavanello A, Previate C, da Silva Silveira S, Ribeiro TA, Martins IP, de Moraes AMP, Matiusso CCI, Saavedra LPJ, de Barros Machado KG, Fabbri Corá T, Gongora A, Cardozo LE, da Silva PHO, Venci R, Vieira E, de Oliveira JC, Miranda RA, de Souza HM, Miksza D, da Costa Lima LD, de Castro-Prado MAA, Rinaldi W, de Freitas Mathias PC. Moderate exercise training since adolescence reduces Walker 256 tumour growth in adult rats. J Physiol 2019; 597:3905-3925. [PMID: 31210356 DOI: 10.1113/jp277645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 05/07/2019] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Cancer growth, cell proliferation and cachexia index can be attenuated by the beneficial programming effect of moderate exercise training, especially if it begins in adolescence. Walker 256 tumour-bearing rats who started exercise training during adolescence did not revert the basal low glycaemia and insulinaemia observed before tumour cell inoculation. The moderate exercise training improved glucose tolerance and peripheral insulin sensitivity only in rats exercised early in adolescence. The chronic effects of our exercise protocol are be beneficial to prevent cancer cachexia and hold clear potential as a nonpharmacological therapy of insulin sensitization. ABSTRACT We tested the hypothesis that moderate exercise training, performed early, starting during adolescence or later in life during adulthood, can inhibit tumour cell growth as a result of changes in biometric and metabolic markers. Male rats that were 30 and 70 days old performed a treadmill running protocol over 8 weeks for 3 days week-1 , 44 min day-1 and at 55-65% V ̇ O 2 max . After the end of training, a batch of rats was inoculated with Walker 256 carcinoma cells. At 15 days after carcinoma cell inoculation, the tumour was weighed and certain metabolic parameters were evaluated. The data demonstrated that physical performance was better in rats that started exercise training during adolescence according to the final workload and V ̇ O 2 max . Early or later moderate exercise training decreased the cachexia index, cell proliferation and tumour growth; however, the effects were more pronounced in rats that exercised during adolescence. Low glycaemia, insulinaemia and tissue insulin sensitivity was not reverted in Walker 256 tumour-bearing rats who trained during adolescence. Cancer growth can be attenuated by the beneficial programming effect of moderate exercise training, especially if it begins during adolescence. In addition, improvement in glucose-insulin homeostasis might be involved in this process.
Collapse
Affiliation(s)
- Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Douglas Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | | | | | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Carina Previate
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Sandra da Silva Silveira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Ana Maria Praxedes de Moraes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Camila Cristina Ianoni Matiusso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Katia Gama de Barros Machado
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Thauany Fabbri Corá
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Adriane Gongora
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Lucas Eduardo Cardozo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Paulo Henrique Olivieri da Silva
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil.,Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Renan Venci
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Elaine Vieira
- Post-Graduate Program of Physical Education, Catholic University of Brasília, Águas Claras, DF, Brazil
| | | | - Rosiane Aparecida Miranda
- Laboratory of Molecular Endocrinology, Carlos Chagas Filho Biophysis Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Daniele Miksza
- Department of Physiology, State University of Londrina, Londrina, PR, Brazil
| | - Luiz Delmar da Costa Lima
- Superior School of Physical Education and Physical Therapy of Goiás State, State University of Goiás, Goiânia, GO, Brazil
| | - Marialba Avezum Alves de Castro-Prado
- Laboratory of Microorganisms Genetics and Mutagenesis, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| | - Wilson Rinaldi
- Department of Physical Education, State University of Maringá, Maringá, PR, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, PR, Brazil
| |
Collapse
|
23
|
Marin BK, Lima Reis SR, Fátima Silva Ramalho A, Lemes SF, Marin L, Vanzela EC, Boschero AC, Carneiro EM, Latorraca MQ, Arantes VC, Barros Reis MA. Protein restriction in early life increases intracellular calcium and insulin secretion, but does not alter expression of SNARE proteins during pregnancy. Exp Physiol 2019; 104:1029-1037. [DOI: 10.1113/ep087045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/24/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Bianca Karine Marin
- Mestrado em Biociências, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | - Silvia Regina Lima Reis
- Departamento de Alimentos Nutrição, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | | | - Simone Ferreira Lemes
- Mestrado em Biociências, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | - Leonardo Marin
- Mestrado em Biociências, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | - Emerielle Cristine Vanzela
- Departamento de Anatomia, Biologia Celular, Fisiologia e BiofísicaInstituto de Biologia, Universidade Estadual de Campinas, Avenida Bertrand Russel, s/n, Cidade Universitária Zeferino Vaz Campinas SP Brazil
| | - Antonio Carlos Boschero
- Departamento de Anatomia, Biologia Celular, Fisiologia e BiofísicaInstituto de Biologia, Universidade Estadual de Campinas, Avenida Bertrand Russel, s/n, Cidade Universitária Zeferino Vaz Campinas SP Brazil
| | - Everardo Magalhães Carneiro
- Departamento de Anatomia, Biologia Celular, Fisiologia e BiofísicaInstituto de Biologia, Universidade Estadual de Campinas, Avenida Bertrand Russel, s/n, Cidade Universitária Zeferino Vaz Campinas SP Brazil
| | - Márcia Queiroz Latorraca
- Departamento de Alimentos Nutrição, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | - Vanessa Cristina Arantes
- Departamento de Alimentos Nutrição, Faculdade de NutriçãoUniversidade Federal de Mato Grosso Cuiabá MT Brazil
| | | |
Collapse
|
24
|
Marin L, Silva HBF, Damin G, Ignacio-Souza LM, Reis SRDL, de Oliveira CAM, Ribeiro RA, Reis MADB, Latorraca MQ, Ferreira F, Arantes VC. Nutritional recovery from a low-protein diet during pregnancy does not restore the kinetics of insulin secretion and Ca 2+ or alterations in the cAMP/PKA and PLC/PKC pathways in islets from adult rats. Appl Physiol Nutr Metab 2019; 43:1257-1267. [PMID: 29758169 DOI: 10.1139/apnm-2017-0629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated the insulin release induced by glucose, the Ca2+ oscillatory pattern, and the cyclic AMP (cAMP)/protein kinase A (PKA) and phospholipase C (PLC)/protein kinase C (PKC) pathways in islets from adult rats that were reared under diets with 17% protein (C) or 6% protein (LP) during gestation, suckling, and after weaning and in rats receiving diets with 6% protein during gestation and 17% protein after birth (R). First-phase glucose-induced insulin secretion was reduced in LP and R islets, and the second phase was partially restored in the R group. Glucose stimulation did not modify intracellular Ca2+ concentration, but it reduced the Ca2+ oscillatory frequency in the R group compared with the C group. Intracellular cAMP concentration was higher and PKA-Cα expression was lower in the R and LP groups compared with the C group. The PKCα content in islets from R rats was lower than that in C and LP rats. Thus, nutritional recovery from a low-protein diet during fetal life did not repair the kinetics of insulin release, impaired Ca2+ handling, and altered the cAMP/PKA and PLC/PKC pathways.
Collapse
Affiliation(s)
- Leonardo Marin
- a Faculdade de Nutrição, Universidade Federal de Mato Grosso, Cuiabá-MT, 78060-900, Brazil
| | | | - Gabriela Damin
- a Faculdade de Nutrição, Universidade Federal de Mato Grosso, Cuiabá-MT, 78060-900, Brazil
| | | | | | | | - Rosane Aparecida Ribeiro
- d Universidade Federal do Rio de Janeiro, Núcleo em Ecologia e desenvolvimento socioambiental, Rio de Janeiro-RJ, 21941-901, Brazil
| | | | | | - Fabiano Ferreira
- e Departamento de Fisiologia e Farmacologia da Universidade Federal de Pernambuco, Recife-PE, 50670-901, Brazil
| | | |
Collapse
|
25
|
Mateus Gonçalves L, Vettorazzi JF, Vanzela EC, Figueiredo MS, Batista TM, Zoppi CC, Boschero AC, Carneiro EM. Amino acid restriction increases β-cell death under challenging conditions. J Cell Physiol 2019; 234:16679-16684. [PMID: 30815898 DOI: 10.1002/jcp.28389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 11/08/2022]
Abstract
Malnutrition programs metabolism, favor dysfunction of β cells. We aimed to establish an in vitro protocol of malnutrition, assessing the effect of amino acid restriction upon the β cells. Insulin-producing cells INS-1E and pancreatic islets were maintained in RPMI 1640 medium containing 1× (Ctl) or 0.25× (AaR) of amino acids. We evaluated several markers of β-cell function and viability. AaR Insulin secretion was reduced, whereas cell viability was unaltered. Calcium oscillations in response to glucose increased in AaR. AaR showed lower Ins1 RNAm, snap 25, and PKC (protein kinase C) protein content, whereas phospho-eIF2α was increased. AaR cells exposed to nutrient or chemical challenges displayed higher apoptosis rates. We showed that amino acid restriction programmed β cell and induced functional changes. This model might be useful for the study of molecular mechanisms involved with β-cell programming helping to establish novel therapeutic targets to prevent harmful outcomes of malnutrition.
Collapse
Affiliation(s)
- Luciana Mateus Gonçalves
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Jean Franciesco Vettorazzi
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Emerielle Cristine Vanzela
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Mariana Sarto Figueiredo
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Thiago Martins Batista
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Claudio Cesar Zoppi
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Antonio Carlos Boschero
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| | - Everardo Magalhães Carneiro
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center (OCRC), Institute of Biology, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
26
|
Lorza‐Gil E, de Souza JC, García‐Arévalo M, Vettorazzi JF, Marques AC, Salerno AG, Trigo JR, Oliveira HCF. Coenzyme Q
10
protects against β‐cell toxicity induced by pravastatin treatment of hypercholesterolemia. J Cell Physiol 2018; 234:11047-11059. [DOI: 10.1002/jcp.27932] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/25/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Estela Lorza‐Gil
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Jane C. de Souza
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Marta García‐Arévalo
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Jean F. Vettorazzi
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Ana Carolina Marques
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Alessandro G. Salerno
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Jose Roberto Trigo
- Department of Animal Biology Biology Institute, State University of Campinas Campinas SP Brazil
| | - Helena C. F. Oliveira
- Department of Structural and Functional Biology Biology Institute, State University of Campinas Campinas SP Brazil
| |
Collapse
|
27
|
Venci RDO, Ramos GB, Martins IP, Matiusso CCI, Saavedra LPJ, Ribeiro TA, Pavanello A, Prates KV, Tófolo LP, Moraes AMPD, Fabricio GS, de Oliveira JC, Franco CCDS, Palma-Rigo K, Mathias PCDF, Malta A. Malnutrition during late pregnancy exacerbates high-fat-diet-induced metabolic dysfunction associated with lower sympathetic nerve tonus in adult rat offspring. Nutr Neurosci 2018; 23:432-443. [DOI: 10.1080/1028415x.2018.1516845] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Renan de Oliveira Venci
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Gabriel Bortoli Ramos
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Camila Cristina Ianoni Matiusso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Ana Maria Praxedes de Moraes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Gabriel Sergio Fabricio
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | | | | | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| |
Collapse
|
28
|
Moreira GV, Azevedo FF, Ribeiro LM, Santos A, Guadagnini D, Gama P, Liberti EA, Saad M, Carvalho C. Liraglutide modulates gut microbiota and reduces NAFLD in obese mice. J Nutr Biochem 2018; 62:143-154. [PMID: 30292107 DOI: 10.1016/j.jnutbio.2018.07.009] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/19/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023]
Abstract
Metabolic disorders such as insulin resistance and diabetes are associated with obesity and nonalcoholic fatty liver disease (NAFLD). The aggressive form of a fatty liver disease may progress to cirrhosis and hepatocellular carcinoma. Furthermore, recent studies demonstrated that there is a dysbiosis in the gut microbiota associated with early stages of metabolic disease. Therefore, the identification and repurposing of drugs already used to treat insulin resistance may be an excellent option for other disorders. We evaluated the effect of liraglutide on obesity, NAFLD and gut microbiota modulation in two different animal models of obesity: the ob/ob mice and the high-fat diet (HFD)-fed mice. Liraglutide treatment induced significant weight loss in both obesity models, showed improvements in glycemic parameters and reduced inflammatory cell infiltration in the cecum and the liver. In ob/ob mice, the liraglutide treatment was able to reduce the accumulation of liver fat by 78% and reversed steatosis in the HFD mice. The gut microbiota analysis showed that liraglutide changed the overall composition as well as the relative abundance of weight-relevant phylotypes such as a reduction of Proteobacteria and an increase of Akkermansia muciniphila in the treated HFD group. We show that liraglutide can lead to weight loss and gut microbiota modulations, and is associated with an improvement of NAFLD. Furthermore, by generating a profile of the intestinal microbiota, we compiled a list of potential bacterial targets that may modulate metabolism and induce a metabolic profile that is considered normal or clinically controlled.
Collapse
Affiliation(s)
- G V Moreira
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical
| | - F F Azevedo
- State University of Campinas-School of Nursing
| | - L M Ribeiro
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical
| | - A Santos
- Department of Internal Medicine, State University of Campinas
| | - D Guadagnini
- Department of Internal Medicine, State University of Campinas
| | - P Gama
- Institute Biomedical Sciences, University of Sao Paulo-Department of Cell and Developmental Biology
| | - E A Liberti
- Institute Biomedical Sciences, University of Sao Paulo-Department of Anatomy
| | - Mja Saad
- Department of Internal Medicine, State University of Campinas
| | - Cro Carvalho
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical.
| |
Collapse
|
29
|
Protein-restriction diet during the suckling phase programs rat metabolism against obesity and insulin resistance exacerbation induced by a high-fat diet in adulthood. J Nutr Biochem 2018; 57:153-161. [DOI: 10.1016/j.jnutbio.2018.03.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 02/09/2018] [Accepted: 03/07/2018] [Indexed: 01/13/2023]
|
30
|
Muñoz VR, Gaspar RC, Crisol BM, Formigari GP, Sant'Ana MR, Botezelli JD, Gaspar RS, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. Physical exercise reduces pyruvate carboxylase (PCB) and contributes to hyperglycemia reduction in obese mice. J Physiol Sci 2018; 68:493-501. [PMID: 28710665 PMCID: PMC10717866 DOI: 10.1007/s12576-017-0559-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/25/2017] [Indexed: 01/07/2023]
Abstract
The present study evaluated the effects of exercise training on pyruvate carboxylase protein (PCB) levels in hepatic tissue and glucose homeostasis control in obese mice. Swiss mice were distributed into three groups: control mice (CTL), fed a standard rodent chow; diet-induced obesity (DIO), fed an obesity-inducing diet; and a third group, which also received an obesity-inducing diet, but was subjected to an exercise training protocol (DIO + EXE). Protocol training was carried out for 1 h/d, 5 d/wk, for 8 weeks, performed at an intensity of 60% of exhaustion velocity. An insulin tolerance test (ITT) was performed in the last experimental week. Twenty-four hours after the last physical exercise session, the animals were euthanized and the liver was harvested for molecular analysis. Firstly, DIO mice showed increased epididymal fat and serum glucose and these results were accompanied by increased PCB and decreased p-Akt in hepatic tissue. On the other hand, physical exercise was able to increase the performance of the mice and attenuate PCB levels and hyperglycemia in DIO + EXE mice. The above findings show that physical exercise seems to be able to regulate hyperglycemia in obese mice, suggesting the participation of PCB, which was enhanced in the obese condition and attenuated after a treadmill running protocol. This is the first study to be aimed at the role of exercise training in hepatic PCB levels, which may be a novel mechanism that can collaborate to reduce the development of hyperglycemia and type 2 diabetes in DIO mice.
Collapse
Affiliation(s)
- Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Barbara Moreira Crisol
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | | | - Marcella Ramos Sant'Ana
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Diego Botezelli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rodrigo Stellzer Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
- OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
- CEPECE-Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.
| |
Collapse
|
31
|
da Rosa CVD, de Campos JM, de Sá Nakanishi AB, Comar JF, Martins IP, Mathias PCDF, Pedrosa MMD, de Godoi VAF, Natali MRM. Food restriction promotes damage reduction in rat models of type 2 diabetes mellitus. PLoS One 2018; 13:e0199479. [PMID: 29924854 PMCID: PMC6010257 DOI: 10.1371/journal.pone.0199479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/07/2018] [Indexed: 12/27/2022] Open
Abstract
There are several animal models of type 2 diabetes mellitus induction but the comparison between models is scarce. Food restriction generates benefits, such as reducing oxidative stress, but there are few studies on its effects on diabetes. The objective of this study is to evaluate the differences in physiological and biochemical parameters between diabetes models and their responses to food restriction. For this, 30 male Wistar rats were distributed in 3 groups (n = 10/group): control (C); diabetes with streptozotocin and cafeteria-style diet (DE); and diabetes with streptozotocin and nicotinamide (DN), all treated for two months (pre-food restriction period). Then, the 3 groups were subdivided into 6, generating the groups CC (control), CCR (control+food restriction), DEC (diabetic+standard diet), DER (diabetic+food restriction), DNC (diabetic+standard diet) and DNR (diabetic+food restriction), treated for an additional two months (food restriction period). The food restriction (FR) used was 50% of the average daily dietary intake of group C. Throughout the treatment, physiological and biochemical parameters were evaluated. At the end of the treatment, serum biochemical parameters, oxidative stress and insulin were evaluated. Both diabetic models produced hyperglycemia, polyphagia, polydipsia, insulin resistance, high fructosamine, hepatic damage and reduced insulin, although only DE presented human diabetes-like alterations, such as dyslipidemia and neuropathy symptoms. Both DEC and DNC diabetic groups presented higher levels of protein carbonyl groups associated to lower antioxidant capacity in the plasma. FR promoted improvement of glycemia in DNR, lipid profile in DER, and insulin resistance and hepatic damage in both diabetes models. FR also reduced the protein carbonyl groups of both DER and DNR diabetic groups, but the antioxidant capacity was improved only in the plasma of DER group. It is concluded that FR is beneficial for diabetes but should be used in conjunction with other therapies.
Collapse
Affiliation(s)
| | | | | | | | - Isabela Peixoto Martins
- Department of Biotechnology, Cell Biology and Genetics State University of Maringá, Paraná, Brazil
| | | | | | | | | |
Collapse
|
32
|
Moreira VM, da Silva Franco CC, Prates KV, Gomes RM, de Moraes AMP, Ribeiro TA, Martins IP, Previate C, Pavanello A, Matiusso CCI, Almeida DL, Francisco FA, Malta A, Tófolo LP, da Silva Silveira S, Saavedra LPJ, Machado K, da Silva PHO, Fabrício GS, Palma-Rigo K, de Souza HM, de Fátima Silva F, Biazi GR, Pereira TS, Vieira E, Miranda RA, de Oliveira JC, da Costa Lima LD, Rinaldi W, Ravanelli MI, de Freitas Mathias PC. Aerobic Exercise Training Attenuates Tumor Growth and Reduces Insulin Secretion in Walker 256 Tumor-Bearing Rats. Front Physiol 2018; 9:465. [PMID: 29867528 PMCID: PMC5953341 DOI: 10.3389/fphys.2018.00465] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 04/13/2018] [Indexed: 12/19/2022] Open
Abstract
Aerobic exercise training can improve insulin sensitivity in many tissues; however, the relationship among exercise, insulin, and cancer cell growth is unclear. We tested the hypothesis that aerobic exercise training begun during adolescence can attenuate Walker 256 tumor growth in adult rats and alter insulin secretion. Thirty-day-old male Wistar rats engaged in treadmill running for 8 weeks, 3 days/week, 44 min/day, at 55-65% VO2max until they were 90 days old (TC, Trained Control). An equivalently aged group was kept inactive during the same period (SC, Sedentary Control). Then, half the animals of the SC and TC groups were reserved as the control condition and the other half were inoculated with Walker 256 cancer cells, yielding two additional groups (Sedentary Walker and Trained Walker). Zero mortalities were observed in tumor-bearing rats. Body weight (BW), food intake, plasma glucose, insulin levels, and peripheral insulin sensitivity were analyzed before and after tumor cell inoculation. We also evaluated tumor growth, metastasis and cachexia. Isolated pancreatic islets secretory activity was analyzed. In addition, we evaluated mechanic sensibility. Our results showed improved physical performance according to the final workload and VO2max and reduced BW in trained rats at the end of the running protocol. Chronic adaptation to the aerobic exercise training decreased tumor weight, cachexia and metastasis and were associated with low glucose and insulin levels and high insulin sensitivity before and after tumor cell inoculation. Aerobic exercise started at young age also reduced pancreatic islet insulin content and insulin secretion in response to a glucose stimulus, without impairing islet morphology in trained rats. Walker 256 tumor-bearing sedentary rats also presented reduced pancreatic islet insulin content, without changing insulin secretion through isolated pancreatic islets. The mechanical sensitivity test indicated that aerobic exercise training did not cause injury or trigger inflammatory processes prior to tumor cell inoculation. Taken together, the current study suggests that aerobic exercise training applied during adolescence may mitigate tumor growth and related disorders in Walker 256 tumor-bearing adult rats. Improved insulin sensibility, lower glucose and insulin levels and/or reduced insulin secretion stimulated by glucose may be implicated in this tumor attenuation.
Collapse
Affiliation(s)
- Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
- Department of Physical Education, State University of Maringá, Maringá, Brazil
- Department of Physical Education, Ingá University Center, UNINGÁ, Maringá, Brazil
| | | | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Rodrigo Mello Gomes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Ana Maria Praxedes de Moraes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Carina Previate
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Camila Cristina Ianoni Matiusso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Douglas Lopes Almeida
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
- Department of Physical Education, State University of Maringá, Maringá, Brazil
- Department of Physical Education, Biomedical Sciences Faculty of Cacoal, Cacoal, Brazil
| | - Sandra da Silva Silveira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Lucas Paulo Jacinto Saavedra
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Katia Machado
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Paulo Henrique Olivieri da Silva
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
- Department of Physical Education, State University of Maringá, Maringá, Brazil
| | - Gabriel S. Fabrício
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | | | | | | | - Taís Susane Pereira
- Laboratory of Microorganisms Genetics and Mutagenesis, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| | - Elaine Vieira
- Post-Graduate Program of Physical Education, Catholic University of Brasília, Águas Claras, Brazil
| | - Rosiane Aparecida Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Luiz Delmar da Costa Lima
- Superior School of Physical Education and Physical Therapy of Goiás State, State University of Goiás, Goiânia, Brazil
| | - Wilson Rinaldi
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
- Department of Physical Education, State University of Maringá, Maringá, Brazil
| | | | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Brazil
| |
Collapse
|
33
|
Piovan S, Pavanello A, Peixoto GML, Matiusso CCI, de Moraes AMP, Martins IP, Malta A, Palma-Rigo K, da Silva Franco CC, Milani PG, Dacome AS, da Costa SC, de Freitas Mathias PC, Mareze-Costa CE. Stevia Nonsweetener Fraction Displays an Insulinotropic Effect Involving Neurotransmission in Pancreatic Islets. Int J Endocrinol 2018; 2018:3189879. [PMID: 29853880 PMCID: PMC5949184 DOI: 10.1155/2018/3189879] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/26/2018] [Indexed: 12/22/2022] Open
Abstract
Stevia rebaudiana (Bert.) Bertoni besides being a source of noncaloric sweeteners is also an important source of bioactive molecules. Many plant extracts, mostly obtained with ethyl acetate solvent, are rich in polyphenol compounds that present insulinotropic effects. To investigate whether the nonsweetener fraction, which is rich in phenolic compounds isolated from Stevia rebaudiana with the solvent ethyl acetate (EAF), has an insulinotropic effect, including interference at the terminals of the autonomic nervous system of the pancreatic islets of rats. Pancreatic islets were isolated from Wistar rats and incubated with EAF and inhibitory or stimulatory substances of insulin secretion, including cholinergic and adrenergic agonists and antagonists. EAF potentiates glucose-stimulated insulin secretion (GSIS) only in the presence of high glucose and calcium-dependent concentrations. EAF increased muscarinic insulinotropic effects in pancreatic islets, interfering with the muscarinic receptor subfamily M3. Adrenergic inhibitory effects on GSIS were attenuated in the presence of EAF, which interfered with the adrenergic α2 receptor. Results suggest that EAF isolated from stevia leaves is a potential therapy for treating type 2 diabetes mellitus by stimulating insulin secretion only in high glucose concentrations, enhancing parasympathetic signal transduction and inhibiting sympathetic signal transduction in beta cells.
Collapse
Affiliation(s)
- Silvano Piovan
- Department of Physiology Sciences, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - Audrei Pavanello
- Department of Cell Biology and Genetics, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | | | | | | | - Isabela Peixoto Martins
- Department of Cell Biology and Genetics, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - Ananda Malta
- Department of Cell Biology and Genetics, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | - Kesia Palma-Rigo
- Department of Cell Biology and Genetics, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | | | - Paula Gimenez Milani
- Department of Biochemistry, Universidade Estadual de Maringá, Maringá, PR, Brazil
| | | | | | | | | |
Collapse
|
34
|
Soares GM, Zangerolamo L, Azevedo EG, Costa-Júnior JM, Carneiro EM, Saad ST, Boschero AC, Barbosa-Sampaio HC. Whole body ARHGAP21 reduction improves glucose homeostasis in high-fat diet obese mice. J Cell Physiol 2018; 233:7112-7119. [PMID: 29574752 DOI: 10.1002/jcp.26527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/31/2018] [Indexed: 12/11/2022]
Abstract
GTPase activating proteins (GAPs) are ubiquitously expressed, and their role in cellular adhesion and membrane traffic processes have been well described. TBC1D1, which is a Rab-GAP, is necessary for adequate glucose uptake by muscle cells, whereas increased TCGAP, which is a Rho-GAP, decreases GLUT4 translocation, and consequently glucose uptake in adipocytes. Here, we assessed the possible involvement of ARHGAP21, a Rho-GAP protein, in glucose homeostasis. For this purpose, wild type mice and ARHGAP21 transgenic whole-body gene-deficiency mice (heterozygous mice, expressing approximately 50% of ARHGAP21) were fed either chow (Ctl and Het) or high-fat diet (Ctl-HFD and Het-HFD). Het-HFD mice showed a reduction in white fat storage, reflected in a lower body weight gain. These mice also displayed an improvement in insulin sensitivity and glucose tolerance, which likely contributed to reduced insulin secretion and pancreatic beta cell area. The reduction of body weight was also observed in Het mice and this phenomenon was associated with an increase in brown adipose tissue and reduced muscle weight, without alteration in glucose-insulin homeostasis. In conclusion, the whole body ARHGAP21 reduction improved glucose homeostasis and protected against diet-induced obesity specifically in Het-HFD mice. However, the mechanism by which ARHGAP21 leads to these outcomes requires further investigation.
Collapse
Affiliation(s)
- Gabriela M Soares
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Lucas Zangerolamo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Elis G Azevedo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Jose M Costa-Júnior
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Everardo M Carneiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Sara T Saad
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO-UNICAMP, Campinas, São Paulo, Brazil
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Helena C Barbosa-Sampaio
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| |
Collapse
|
35
|
Muñoz VR, Gaspar RC, Kuga GK, Nakandakari SCBR, Baptista IL, Mekary RA, da Silva ASR, de Moura LP, Ropelle ER, Cintra DE, Pauli JR. Exercise decreases CLK2 in the liver of obese mice and prevents hepatic fat accumulation. J Cell Biochem 2018; 119:5885-5892. [PMID: 29575149 DOI: 10.1002/jcb.26780] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 02/02/2018] [Indexed: 12/31/2022]
Abstract
The accumulation of fatty acids in the liver associated with obesity condition is also known as nonalcoholic fatty liver disease (NAFLD). The impaired fat oxidation in obesity condition leads to increased hepatic fat accumulation and increased metabolic syndrome risk. On the other hand, physical exercise has been demonstrated as a potent strategy in the prevention of NAFLD. Also, these beneficial effects of exercise occur through different mechanisms. Recently, the Cdc2-like kinase (CLK2) protein was associated with the suppression of fatty acid oxidation and hepatic ketogenesis. Thus, obese animals demonstrated elevated levels of hepatic CLK2 and decreased fat acid oxidation. Here, we explored the effects of chronic physical exercise in the hepatic metabolism of obese mice. Swiss mice were distributed in Lean, Obese (fed with high-fat diet during 16 weeks) and Trained Obese group (fed with high-fat diet during 16 weeks and exercised (at 60% exhaustion velocity during 1 h/5 days/week) during 8 weeks. In our results, the obese animals showed insulin resistance, increased hepatic CLK2 content and increased hepatic fat accumulation compared to the Lean group. Otherwise, the chronic physical exercise improved insulin resistance state, prevented the increased CLK2 in the liver and attenuated hepatic fat accumulation. In summary, these data reveal a new protein involved in the prevention of hepatic fat accumulation after chronic physical exercise. More studies can evidence the negative role of CLK2 in the control of liver metabolism, contributing to the improvement of insulin resistance, obesity, and type 2 diabetes.
Collapse
Affiliation(s)
- Vitor R Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael C Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Gabriel K Kuga
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Susana C B R Nakandakari
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Igor L Baptista
- Laboratory of Cell and Tissue Biology, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rania A Mekary
- Department of Nutrition, Harvard T. Chan School of Public Health, Boston, Massachusetts.,Department of Social and Administrative Sciences, MCPHS University, Boston, Massachusetts
| | - Adelino S R da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Leandro P de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo R Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José R Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC-Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,CEPECE-Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
36
|
Prates KV, de Oliveira JC, Malta A, Matiusso CCI, Miranda RA, Ribeiro TA, Francisco FA, Franco CCS, Moreira VM, Alves VS, Torrezan R, Mathias PCF, Barella LF. Sympathetic innervation is essential for metabolic homeostasis and pancreatic beta cell function in adult rats. Mol Cell Endocrinol 2018; 462:119-126. [PMID: 28962894 DOI: 10.1016/j.mce.2017.09.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 11/16/2022]
Abstract
Obesity is associated with an imbalance in the activity of the autonomic nervous system (ANS), specifically in the organs involved in energy metabolism. The pancreatic islets are richly innervated by the ANS, which tunes the insulin release due to changes in energy demand. Therefore, changes in the sympathetic input that reach the pancreas can lead to metabolic dysfunctions. To evaluate the role of the sympathetic ends that innervate the pancreas, 60-day-old male Wistar rats were subjected to sympathectomy (SYM) or were sham-operated (SO). At 120 day-old SYM rats exhibited an increase in body weight, fat pads and metabolic dysfunctions. Decreases in the HOMA-IR and reductions in insulin release were observed both in vivo and in vitro. Furthermore, the SYM rats exhibited altered pancreatic islet function in both muscarinic and adrenergic assays and exhibited high protein expression of the alpha-2 adrenergic receptor (α2AR). Because α2AR has been linked to type 2 diabetes, these findings demonstrate the clinical implications of this study.
Collapse
Affiliation(s)
- Kelly V Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil.
| | - Júlio C de Oliveira
- Institute of Health Sciences, Federal University of Mato Grosso, Sinop, MT, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Camila C I Matiusso
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Rosiane A Miranda
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tatiane A Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Flávio A Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Claudinéia C S Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Veridiana M Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Vander S Alves
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Rosana Torrezan
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Paulo C F Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| | - Luiz F Barella
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá, Maringá, Paraná, Brazil
| |
Collapse
|
37
|
Besson JCF, Hernandes L, Campos JMD, Morikawa KA, Bersani-Amado CA, Matioli G. Insulin complexed with cyclodextrins stimulates epithelialization and neovascularization of skin wound healing in rats. Injury 2017; 48:2417-2425. [PMID: 28888719 DOI: 10.1016/j.injury.2017.08.046] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 08/21/2017] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Skin lesions are a significant public health problem, above all that wounds fail to heal properly and become chronic. Due to its reepithelization action, insulin has the potential to heal skin lesions, by stimulating the proliferation and migration of keratinocytes, angiogenic stimulus, and increasing collagen deposition. In the present study insulin was complexed with 2-hydroxypropyl-β-cyclodextrin (HPβCD) and its wound healing effect and inclusion complex (HPβCD-I) were evaluated in excisional wounds in the skin of rats. MATERIAL AND METHODS Three different gel based pharmaceutical forms were created: carbopol 940® base gel, an insulin gel comprising the base gel plus 50 IU of insulin and a gel complex comprising the base gel plus (HPβCD) complexed with insulin (HPβCD-I) were used to verify wound healing in vitro and in vivo assays. RESULTS The wounds in the skin of rats were treated with gel containing HPβCD-I not cytoxically irritating and cytotoxic. Analysis of cell proliferation and measurement of the length and thickness of the epidermis showed that HPβCD-I prolonged the proliferation and migration of keratinocytes. Revascularization analysis of lesions treated with HPβCD-I compared to those treated with insulin found that angiogenic stimulus was less intense, but more constant and prolonged in the modified release process. There was increased deposition of type I and III collagen fibers in accordance with the treatment time. CONCLUSION Therefore, the slow release of complexed insulin modulated the reepithelialization process by stimulating cell proliferation and migration of keratinocytes, favoring greater concentration of serum insulin, modulating inflammatory response, matrix remodeling and promoting neovascularization. Angiogenesis extended by the steady release of insulin can be effective in the treatment of chronic wounds.
Collapse
Affiliation(s)
- Jean Carlos Fernando Besson
- Department of Morphologic Sciences, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil
| | - Luzmarina Hernandes
- Department of Morphologic Sciences, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil
| | - Jéssica Men de Campos
- Department of Morphologic Sciences, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil
| | - Karina Amélia Morikawa
- Department of Morphologic Sciences, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil
| | - Ciomar Aparecida Bersani-Amado
- Department of Pharmacology and Therapeutics, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil
| | - Graciette Matioli
- Department of Pharmacy, State University of Maringá, Av. Colombo, 5790-87020-900 Maringá, PR, Brazil.
| |
Collapse
|
38
|
Cappelli APG, Zoppi CC, Silveira LR, Batista TM, Paula FM, da Silva PMR, Rafacho A, Barbosa-Sampaio HC, Boschero AC, Carneiro EM. Reduced glucose-induced insulin secretion in low-protein-fed rats is associated with altered pancreatic islets redox status. J Cell Physiol 2017; 233:486-496. [PMID: 28370189 DOI: 10.1002/jcp.25908] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/13/2017] [Indexed: 01/01/2023]
Abstract
In the present study, we investigated the relationship between early life protein malnutrition-induced redox imbalance, and reduced glucose-stimulated insulin secretion. After weaning, male Wistar rats were submitted to a normal-protein-diet (17%-protein, NP) or to a low-protein-diet (6%-protein, LP) for 60 days. Pancreatic islets were isolated and hydrogen peroxide (H2 O2 ), oxidized (GSSG) and reduced (GSH) glutathione content, CuZn-superoxide dismutase (SOD1), glutathione peroxidase (GPx1) and catalase (CAT) gene expression, as well as enzymatic antioxidant activities were quantified. Islets that were pre-incubated with H2 O2 and/or N-acetylcysteine, were subsequently incubated with glucose for insulin secretion measurement. Protein malnutrition increased CAT mRNA content by 100%. LP group SOD1 and CAT activities were 50% increased and reduced, respectively. H2 O2 production was more than 50% increased whereas GSH/GSSG ratio was near 60% lower in LP group. Insulin secretion was, in most conditions, approximately 50% lower in LP rat islets. When islets were pre-incubated with H2 O2 (100 μM), and incubated with glucose (33 mM), LP rats showed significant decrease of insulin secretion. This effect was attenuated when LP islets were exposed to N-acetylcysteine.
Collapse
Affiliation(s)
- Ana Paula G Cappelli
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão (UFMA), São Luís, Maranhão, Brazil.,Department of Physiology and Biophysiology, Institute of Biomedical Sciences, University of Sao Paulo (USP), São Paulo, Brazil
| | - Claudio C Zoppi
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Leonardo R Silveira
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Thiago M Batista
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Flávia M Paula
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | | | - Alex Rafacho
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Department of Physiologic Sciences, Center of Biologic Sciences, Federal University of Santa Catarina (UFSC), Florianopolis, Santa Catarina, Brazil
| | - Helena C Barbosa-Sampaio
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Antonio C Boschero
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Everardo M Carneiro
- Department of Structural and Functional Biology, Cellular Biology and Physiology and Biophysics, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
39
|
miR-124a expression contributes to the monophasic pattern of insulin secretion in islets from pregnant rats submitted to a low-protein diet. Eur J Nutr 2017; 57:1471-1483. [PMID: 28314963 DOI: 10.1007/s00394-017-1425-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 03/01/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE To evaluate the role of miR-124a in the regulation of genes involved in insulin exocytosis and its effects on the kinetics of insulin secretion in pancreatic islets from pregnant rats submitted to a low-protein diet. METHODS Adult control non-pregnant (CNP) and control pregnant (CP) rats were fed a normal protein diet (17%), whereas low-protein non-pregnant (LPNP) and low-protein pregnant (LPP) rats were fed a low-protein diet (6%) from days 1 to 15 of pregnancy. Kinetics of the glucose-induced insulin release and measurement of [Ca2+]i in pancreatic islets were assessed by standard protocols. The miR-124a expression and gene transcriptions from pancreatic islets were determined by real-time polymerase chain reaction. RESULTS In islets from LPP rats, the first phase of insulin release was abrogated. The AUC [Ca2+]i from the LPP group was lower compared with the other groups. miR-124a expression was reduced by a low-protein diet. SNAP-25 mRNA, protein expression, and Rab3A protein content were lower in the LPP rats than in CP rats. Syntaxin 1A and Kir6.2 mRNA levels were decreased in islets from low-protein rats compared with control rats, whereas their protein content was reduced in islets from pregnant rats. CONCLUSIONS Loss of biphasic insulin secretion in islets from LPP rats appears to have resulted from reduced [Ca2+]i due, at least in part, to Kir6.2 underexpression and from the changes in exocytotic elements that are influenced either directly or indirectly by miR-124a.
Collapse
|
40
|
Miranda RA, da Silva Franco CC, de Oliveira JC, Barella LF, Tófolo LP, Ribeiro TA, Pavanello A, da Conceição EPS, Torrezan R, Armitage J, Lisboa PC, de Moura EG, de Freitas Mathias PC, Vieira E. Cross-fostering reduces obesity induced by early exposure to monosodium glutamate in male rats. Endocrine 2017; 55:101-112. [PMID: 27116693 DOI: 10.1007/s12020-016-0965-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 04/16/2016] [Indexed: 12/16/2022]
Abstract
Maternal obesity programmes a range of metabolic disturbances for the offspring later in life. Moreover, environmental changes during the suckling period can influence offspring development. Because both periods significantly affect long-term metabolism, we aimed to study whether cross-fostering during the lactation period was sufficient to rescue a programmed obese phenotype in offspring induced by maternal obesity following monosodium L-glutamate (MSG) treatment. Obesity was induced in female Wistar rats by administering subcutaneous MSG (4 mg/g body weight) for the first 5 days of postnatal life. Control and obese female rats were mated in adulthood. The resultant pups were divided into control second generation (F2) (CTLF2), MSG-treated second generation (F2) (MSGF2), which suckled from their CTL and MSG biological dams, respectively, or CTLF2-CR, control offspring suckled by MSG dams and MSGF2-CR, MSG offspring suckled by CTL dams. At 120 days of age, fat tissue accumulation, lipid profile, hypothalamic leptin signalling, glucose tolerance, glucose-induced, and adrenergic inhibition of insulin secretion in isolated pancreatic islets were analysed. Maternal MSG-induced obesity led to an obese phenotype in male offspring, characterized by hyperinsulinaemia, hyperglycaemia, hyperleptinaemia, dyslipidaemia, and impaired leptin signalling, suggesting central leptin resistance, glucose intolerance, impaired glucose-stimulated, and adrenergic inhibition of insulin secretion. Cross-fostering normalized body weight, food intake, leptin signalling, lipid profiles, and insulinaemia, but not glucose homeostasis or insulin secretion from isolated pancreatic islets. Our findings suggest that alterations during the lactation period can mitigate the development of obesity and prevent the programming of adult diseases.
Collapse
Affiliation(s)
- Rosiane Aparecida Miranda
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil.
- Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Claudinéia Conationi da Silva Franco
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| | | | - Luiz Felipe Barella
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laize Peron Tófolo
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| | - Audrei Pavanello
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| | - Ellen Paula Santos da Conceição
- Department of Physiological Sciences Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Rosana Torrezan
- Department of Physiological Sciences, State University of Maringá, Maringá, PR, Brazil
| | - James Armitage
- School of Medicine (Optometr), Deakin University, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Patrícia Cristina Lisboa
- Department of Physiological Sciences Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto Gaspar de Moura
- Department of Physiological Sciences Roberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| | - Elaine Vieira
- Department of Biotechnology, Cell Biology and Genetics, State University of Maringá/UEM, Block H67, room 19, Colombo Avenue 5790, Maringá, PR, Brazil
| |
Collapse
|
41
|
Ribeiro TA, Prates KV, Pavanello A, Malta A, Tófolo LP, Martins IP, Oliveira JCD, Miranda RA, Gomes RM, Vieira E, Franco CCDS, Barella LF, Francisco FA, Alves VS, Silveira SDS, Moreira VM, Fabricio GS, Palma-Rigo K, Sloboda DM, Mathias PCDF. Acephate exposure during a perinatal life program to type 2 diabetes. Toxicology 2016; 372:12-21. [PMID: 27765684 DOI: 10.1016/j.tox.2016.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/07/2016] [Accepted: 10/15/2016] [Indexed: 01/10/2023]
Abstract
Acephate has been used extensively as an insecticide in agriculture. Its downstream sequelae are associated with hyperglycemia, lipid metabolism dysfunction, DNA damage, and cancer, which are rapidly growing epidemics and which lead to increased morbidity and mortality rates and soaring health-care costs. Developing interventions will require a comprehensive understanding of which excess insecticides during perinatal life can cause insulin resistance and type 2 diabetes. A Wistar rat animal model suggests that acephate exposure during pregnancy and lactation causes alterations in maternal glucose metabolism and programs the offspring to be susceptible to type 2 diabetes at adulthood. Therapeutic approaches based on preventive actions to food contaminated with insecticides during pregnancy and lactation could prevent new cases of type 2 diabetes.
Collapse
Affiliation(s)
- Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil.
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Júlio Cezar de Oliveira
- Institute of Health Sciences, Federal University of Mato Grosso, 78.557-267 Sinop, MT, Brazil
| | - Rosiane Aparecida Miranda
- Laboratory of Molecular Endocrinology, Carlos Chagas Filho Biophysis Institute, Federal University of Rio de Janeiro, 21.941-902 Rio de Janeiro, RJ, Brazil
| | - Rodrigo Mello Gomes
- Laboratory of Neuroscience and Cardiovascular Physiology, Department of Physiological Sciences, Federal University of Goiás, 74690-900 Goiânia, GO, Brazil
| | - Elaine Vieira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Claudinéia Conationi da Silva Franco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Luiz Felipe Barella
- Molecular Signalling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 20892 Bethesda, MD, USA
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Vander Silva Alves
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Sandra da Silva Silveira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Veridiana Mota Moreira
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Gabriel Sergio Fabricio
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, 59000 Lille, France
| | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, Ob/Gyn, and Pediatrics MacMaster University-8S 4L8, Hamilton, Ontario, Canada
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringá-5790, 87020-900 Maringá, PR, Brazil; UPSP-EGEAL Institut Polytechnique LaSalle de Beauvais, BP, 30313-60026 Beauvais Cedex, France
| |
Collapse
|
42
|
Malta A, Souza AAD, Ribeiro TA, Francisco FA, Pavanello A, Prates KV, Tófolo LP, Miranda RA, Oliveira JCD, Martins IP, Previate C, Gomes RM, Franco CCDS, Natali MRM, Palma-Rigo K, Mathias PCDF. Neonatal treatment with scopolamine butylbromide prevents metabolic dysfunction in male rats. Sci Rep 2016; 6:30745. [PMID: 27561682 PMCID: PMC4999897 DOI: 10.1038/srep30745] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/07/2016] [Indexed: 01/17/2023] Open
Abstract
We tested whether treatment with a cholinergic antagonist could reduce insulin levels in early postnatal life and attenuate metabolic dysfunctions induced by early overfeeding in adult male rats. Wistar rats raised in small litters (SLs, 3 pups/dam) and normal litters (NLs, 9 pups/dam) were used in models of early overfeeding and normal feeding, respectively. During the first 12 days of lactation, animals in the SL and NL groups received scopolamine butylbromide (B), while the controls received saline (S) injections. The drug treatment decreased insulin levels in pups from both groups, and as adults, these animals showed improvements in glucose tolerance, insulin sensitivity, vagus nerve activity, fat tissue accretion, insulinemia, leptinemia, body weight gain and food intake. Low glucose and cholinergic insulinotropic effects were observed in pancreatic islets from both groups. Low protein expression was observed for the muscarinic M3 acetylcholine receptor subtype (M3mAChR), although M2mAChR subtype expression was increased in SL-B islets. In addition, beta-cell density was reduced in drug-treated rats. These results indicate that early postnatal scopolamine butylbromide treatment inhibits early overfeeding-induced metabolic dysfunctions in adult rats, which might be caused by insulin decreases during lactation, associated with reduced parasympathetic activity and expression of M3mAChR in pancreatic islets.
Collapse
Affiliation(s)
- Ananda Malta
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Aline Amenencia de Souza
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Tatiane Aparecida Ribeiro
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Flávio Andrade Francisco
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Audrei Pavanello
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Kelly Valério Prates
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Laize Peron Tófolo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Rosiane Aparecida Miranda
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | | | - Isabela Peixoto Martins
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Carina Previate
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Rodrigo Mello Gomes
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | | | | | - Kesia Palma-Rigo
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| | - Paulo Cezar de Freitas Mathias
- Laboratory of Secretion Cell Biology, Department of Biotechnology, Genetics and Cell Biology, State University of Maringa, Maringá, PR, Brazil
| |
Collapse
|
43
|
Miranda RA, Torrezan R, de Oliveira JC, Barella LF, da Silva Franco CC, Lisboa PC, Moura EG, Mathias PCF. HPA axis and vagus nervous function are involved in impaired insulin secretion of MSG-obese rats. J Endocrinol 2016; 230:27-38. [PMID: 27113853 DOI: 10.1530/joe-15-0467] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 04/22/2016] [Indexed: 11/08/2022]
Abstract
Neuroendocrine dysfunctions such as the hyperactivity of the vagus nerve and hypothalamus-pituitary-adrenal (HPA) axis greatly contribute to obesity and hyperinsulinemia; however, little is known about these dysfunctions in the pancreatic β-cells of obese individuals. We used a hypothalamic-obesity model obtained by neonatal treatment with monosodium l-glutamate (MSG) to induce obesity. To assess the role of the HPA axis and vagal tonus in the genesis of hypercorticosteronemia and hyperinsulinemia in an adult MSG-obese rat model, bilateral adrenalectomy (ADX) and subdiaphragmatic vagotomy (VAG) alone or combined surgeries (ADX-VAG) were performed. To study glucose-induced insulin secretion (GIIS) and the cholinergic insulinotropic process, pancreatic islets were incubated with different glucose concentrations with or without oxotremorine-M, a selective agonist of the M3 muscarinic acetylcholine receptor (M3AChR) subtype. Protein expression of M3AChR in pancreatic islets, corticosteronemia, and vagus nerve activity was also evaluated. Surgeries reduced 80% of the body weight gain. Fasting glucose and insulin were reduced both by ADX and ADX-VAG, whereas VAG was only associated with hyperglycemia. The serum insulin post-glucose stimulation was lower in all animals that underwent an operation. Vagal activity was decreased by 50% in ADX rats. In the highest glucose concentration, both surgeries reduced GIIS by 50%, whereas ADX-VAG decreased by 70%. Additionally, M3AChR activity was recovered by the individual surgeries. M3AChR protein expression was reduced by ADX. Both the adrenal gland and vagus nerve contribute to the hyperinsulinemia in the MSG model, although adrenal is more crucial as it appears to modulate parasympathetic activity and M3AChR expression in obesity.
Collapse
Affiliation(s)
- Rosiane A Miranda
- Department of BiotechnologyGenetics and Cell Biology, State University of Maringá, Maringá, Brazil Carlos Chagas Filho Biophysics InstituteFederal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rosana Torrezan
- Department of Physiological SciencesState University of Maringá, Maringá, Brazil
| | - Júlio C de Oliveira
- Institute of Health SciencesFederal University of Mato Grosso, Sinop, Brazil
| | - Luiz F Barella
- Molecular Signalling SectionLaboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Patrícia C Lisboa
- Department of Physiological SciencesRoberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Egberto G Moura
- Department of Physiological SciencesRoberto Alcântara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo C F Mathias
- Department of BiotechnologyGenetics and Cell Biology, State University of Maringá, Maringá, Brazil
| |
Collapse
|
44
|
Munhoz AC, Riva P, Simões D, Curi R, Carpinelli AR. Control of Insulin Secretion by Production of Reactive Oxygen Species: Study Performed in Pancreatic Islets from Fed and 48-Hour Fasted Wistar Rats. PLoS One 2016; 11:e0158166. [PMID: 27362938 PMCID: PMC4928816 DOI: 10.1371/journal.pone.0158166] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/07/2016] [Indexed: 11/19/2022] Open
Abstract
Mitochondria and NADPH oxidase are important sources of reactive oxygen species in particular the superoxide radical (ROS) in pancreatic islets. These molecules derived from molecular oxygen are involved in pancreatic β-cells signaling and control of insulin secretion. We examined the involvement of ROS produced through NADPH oxidase in the leucine- and/or glucose-induced insulin secretion by pancreatic islets from fed or 48-hour fasted rats. Glucose-stimulated insulin secretion (GSIS) in isolated islets was evaluated at low (2.8 mM) or high (16.7 mM) glucose concentrations in the presence or absence of leucine (20 mM) and/or NADPH oxidase inhibitors (VAS2870–20 μM or diphenylene iodonium—DPI—5 μM). ROS production was determined in islets treated with dihydroethidium (DHE) or MitoSOX Red reagent for 20 min and dispersed for fluorescence measurement by flow cytometry. NADPH content variation was examined in INS-1E cells (an insulin secreting cell line) after incubation in the presence of glucose (2.8 or 16.7 mM) and leucine (20 mM). At 2.8 mM glucose, VAS2870 and DPI reduced net ROS production (by 30%) and increased GSIS (by 70%) in a negative correlation manner (r = -0.93). At 16.7 mM glucose or 20 mM leucine, both NADPH oxidase inhibitors did not alter insulin secretion neither net ROS production. Pentose phosphate pathway inhibition by treatment with DHEA (75 μM) at low glucose led to an increase in net ROS production in pancreatic islets from fed rats (by 40%) and induced a marked increase (by 144%) in islets from 48-hour fasted rats. The NADPH/NADP+ ratio was increased when INS-1E cells were exposed to high glucose (by 4.3-fold) or leucine (by 3-fold). In conclusion, increased ROS production through NADPH oxidase prevents the occurrence of hypoglycemia in fasting conditions, however, in the presence of high glucose or high leucine levels, the increased production of NADPH and the consequent enhancement of the activity of the antioxidant defenses mitigate the excess of ROS production and allow the secretory process of insulin to take place.
Collapse
Affiliation(s)
- Ana Cláudia Munhoz
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- * E-mail:
| | - Patrícia Riva
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniel Simões
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Angelo Rafael Carpinelli
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Sanches JR, França LM, Chagas VT, Gaspar RS, Dos Santos KA, Gonçalves LM, Sloboda DM, Holloway AC, Dutra RP, Carneiro EM, Cappelli APG, Paes AMDA. Polyphenol-Rich Extract of Syzygium cumini Leaf Dually Improves Peripheral Insulin Sensitivity and Pancreatic Islet Function in Monosodium L-Glutamate-Induced Obese Rats. Front Pharmacol 2016; 7:48. [PMID: 27014062 PMCID: PMC4785152 DOI: 10.3389/fphar.2016.00048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/22/2016] [Indexed: 12/23/2022] Open
Abstract
Syzygium cumini (L.) Skeels (Myrtaceae) has been traditionally used to treat a number of illnesses. Ethnopharmacological studies have particularly addressed antidiabetic and metabolic-related effects of extracts prepared from its different parts, especially seed, and pulp-fruit, however. there is a lack of studies on phytochemical profile and biological properties of its leaf. As there is considerable interest in bioactive compounds to treat metabolic syndrome and its clustered risk factors, we sought to characterize the metabolic effects of hydroethanolic extract of S. cumini leaf (HESc) on lean and monosodium L-glutamate (MSG)-induced obese rats. HPLC-MS/MS characterization of the HESc polyphenolic profile, at 254 nm, identified 15 compounds pertaining to hydrolysable tannin and flavanol subclasses. At 60 days of age, both groups were randomly assigned to receive HESc (500 mg/kg) or vehicle for 30 days. At the end of treatment, obese+HESc exhibited significantly lower body weight gain, body mass index, and white adipose tissue mass, compared to obese rats receiving vehicle. Obese rats treated with HESc showed a twofold increase in lipolytic activity in the periepididymal fat pad, as well as, brought triglyceride levels in serum, liver and skeletal muscle back to levels close those found in lean animals. Furthermore, HESc also improved hyperinsulinemia and insulin resistance in obese+HESc rats, which resulted in partial reversal of glucose intolerance, as compared to obese rats. HESc had no effect in lean rats. Assessment of ex vivo glucose-stimulated insulin secretion showed HESc potentiated pancreatic function in islets isolated from both lean and obese rats treated with HESc. In addition, HESc (10–1000 μg/mL) increased glucose stimulated insulin secretion from both isolated rat islets and INS-1E β-cells. These data demonstrate that S. cumini leaf improved peripheral insulin sensitivity via stimulating/modulating β-cell insulin release, which was associated with improvements in metabolic outcomes in MSG-induced obese rats.
Collapse
Affiliation(s)
- Jonas R Sanches
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Lucas M França
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Vinicyus T Chagas
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Renato S Gaspar
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Kayque A Dos Santos
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Luciana M Gonçalves
- Laboratory of Endocrine Pancreas and Metabolism, Department of Estructural and Functional Biology, Institute of Biology, State University of Campinas Campinas, Brazil
| | - Deborah M Sloboda
- Departments of Biochemistry, Pediatrics and Obstetrics and Gynecology, McMaster University Hamilton, ON, Canada
| | - Alison C Holloway
- Department of Obstetrics and Gynecology, McMaster University Hamilton, ON, Canada
| | - Richard P Dutra
- Social, Health and Technological Sciences Center, Federal University of Maranhão Imperatriz, Brazil
| | - Everardo M Carneiro
- Laboratory of Endocrine Pancreas and Metabolism, Department of Estructural and Functional Biology, Institute of Biology, State University of Campinas Campinas, Brazil
| | - Ana Paula G Cappelli
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| | - Antonio Marcus de A Paes
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão São Luís, Brazil
| |
Collapse
|
46
|
Lorza-Gil E, Salerno AG, Wanschel ACBA, Vettorazzi JF, Ferreira MS, Rentz T, Catharino RR, Oliveira HCF. Chronic use of pravastatin reduces insulin exocytosis and increases β-cell death in hypercholesterolemic mice. Toxicology 2016; 344-346:42-52. [PMID: 26875785 DOI: 10.1016/j.tox.2015.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/14/2015] [Accepted: 12/22/2015] [Indexed: 02/04/2023]
Abstract
We have previously demonstrated that hypercholesterolemic LDL receptor knockout (LDLr(-/-)) mice secrete less insulin than wild-type mice. Removing cholesterol from isolated islets using methyl-beta-cyclodextrin reversed this defect. In this study, we hypothesized that in vivo treatment of LDLr(-/-) mice with the HMGCoA reductase inhibitor pravastatin would improve glucose-stimulated insulin secretion. Female LDLr(-/-) mice were treated with pravastatin (400mg/L) for 1-3 months. Isolated pancreatic islets were assayed for insulin secretion rates, intracellular calcium oscillations, cholesterol levels, NAD(P)H and SNARE protein levels, apoptosis indicators and lipidomic profile. Two months pravastatin treatment reduced cholesterol levels in plasma, liver and islets by 35%, 25% and 50%, respectively. Contrary to our hypothesis, pravastatin treatment increased fasting and fed plasma levels of glucose and decreased markedly (40%) fed plasma levels of insulin. In addition, ex vivo glucose stimulated insulin secretion was significantly reduced after two and three months (36-48%, p<0.05) of pravastatin treatment. Although reducing insulin secretion and insulinemia, two months pravastatin treatment did not affect glucose tolerance because it improved global insulin sensitivity. Pravastatin induced islet dysfunction was associated with marked reductions of exocytosis-related SNARE proteins (SNAP25, Syntaxin 1A, VAMP2) and increased apoptosis markers (Bax/Bcl2 protein ratio, cleaved caspase-3 and lower NAD(P)H production rates) observed in pancreatic islets from treated mice. In addition, several oxidized phospholipids, tri- and diacylglycerols and the proapoptotic lipid molecule ceramide were identified as markers of pravastatin-treated islets. Cell death and oxidative stress (H2O2 production) were confirmed in insulin secreting INS-1E cells treated with pravastatin. These results indicate that chronic treatment with pravastatin impairs the insulin exocytosis machinery and increases β-cell death. These findings suggest that prolonged use of statins may have a diabetogenic effect.
Collapse
Affiliation(s)
- Estela Lorza-Gil
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Alessandro G Salerno
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Amarylis C B A Wanschel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Jean F Vettorazzi
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Mônica S Ferreira
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | - Thiago Rentz
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Rodrigo R Catharino
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil.
| |
Collapse
|
47
|
Raposo HF, Vanzela EC, Berti JA, Oliveira HCF. Cholesteryl Ester Transfer Protein (CETP) expression does not affect glucose homeostasis and insulin secretion: studies in human CETP transgenic mice. Lipids Health Dis 2016; 15:9. [PMID: 26758205 PMCID: PMC4711172 DOI: 10.1186/s12944-016-0179-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 01/07/2016] [Indexed: 12/26/2022] Open
Abstract
Background Cholesteryl ester transfer protein (CETP) is a plasma protein that mediates the exchange of triglycerides for esterified cholesterol between HDL and apoB-lipoproteins. Previous studies suggest that CETP may modify glucose metabolism in patients or cultured cells. In this study, we tested if stable CETP expression would impair glucose metabolism. Methods We used human CETP transgenic mice and non-transgenic littermate controls (NTg), fed with control or high fat diet, as well as in dyslipidemic background and aging conditions. Assays included glucose and insulin tolerance tests, isolated islets insulin secretion, tissue glucose uptake and adipose tissue GLUT mRNA expression. Results CETP expression did not modify glucose or insulin tolerance in all tested conditions such as chow and high fat diet, adult and aged mice, normo and dyslipidemic backgrounds. Fasting and fed state plasma levels of insulin were not differ in CETP and NTg mice. Direct measurements of isolated pancreatic islet insulin secretion rates induced by glucose (11, 16.7 or 22 mM), KCl (40 mM), and leucine (10 mM) were similar in NTg and CETP mice, indicating that CETP expression did not affect β-cell function in vivo and ex vivo. Glucose uptake by insulin target tissues, measured in vivo using 3H-2-deoxyglucose, showed that CETP expression had no effect on the glucose uptake in liver, muscle, perigonadal, perirenal, subcutaneous and brown adipose tissues. Accordingly, GLUT1 and GLUT4 mRNA in adipose tissue were not affected by CETP. Conclusions In summary, by comparing the in vivo all-or-nothing CETP expressing mouse models, we demonstrated that CETP per se has no impact on the glucose tolerance and tissue uptake, global insulin sensitivity and beta cell insulin secretion rates.
Collapse
Affiliation(s)
- Helena F Raposo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Unicamp - Cidade Universitária Zeferino Vaz. Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Emerielle C Vanzela
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Unicamp - Cidade Universitária Zeferino Vaz. Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil
| | - Jairo A Berti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Unicamp - Cidade Universitária Zeferino Vaz. Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil.,Present address: Department of Physiological Science, State University of Maringa, Maringa, PR, Brazil
| | - Helena C F Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Unicamp - Cidade Universitária Zeferino Vaz. Rua Monteiro Lobato, 255, Campinas, SP, CEP 13083-862, Brazil.
| |
Collapse
|
48
|
Martin JM, Miranda RA, Barella LF, Palma-Rigo K, Alves VS, Fabricio GS, Pavanello A, Franco CCDS, Ribeiro TA, Visentainer JV, Banafé EG, Martin CA, Mathias PCDF, de Oliveira JC. Maternal Diet Supplementation with n-6/n-3 Essential Fatty Acids in a 1.2 : 1.0 Ratio Attenuates Metabolic Dysfunction in MSG-Induced Obese Mice. Int J Endocrinol 2016; 2016:9242319. [PMID: 28050167 PMCID: PMC5165167 DOI: 10.1155/2016/9242319] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/19/2016] [Accepted: 10/04/2016] [Indexed: 12/25/2022] Open
Abstract
Essential polyunsaturated fatty acids (PUFAs) prevent cardiometabolic diseases. We aimed to study whether a diet supplemented with a mixture of n-6/n-3 PUFAs, during perinatal life, attenuates outcomes of long-term metabolic dysfunction in prediabetic and obese mice. Seventy-day-old virgin female mice were mated. From the conception day, dams were fed a diet supplemented with sunflower oil and flaxseed powder (containing an n-6/n-3 PUFAs ratio of 1.2 : 1.0) throughout pregnancy and lactation, while control dams received a commercial diet. Newborn mice were treated with monosodium L-glutamate (MSG, 4 mg g-1 body weight per day) for the first 5 days of age. A batch of weaned pups was sacrificed to quantify the brain and pancreas total lipids; another batch were fed a commercial diet until 90 days of age, where glucose homeostasis and glucose-induced insulin secretion (GIIS) as well as retroperitoneal fat and Lee index were assessed. MSG-treated mice developed obesity, glucose intolerance, insulin resistance, pancreatic islet dysfunction, and higher fat stores. Maternal flaxseed diet-supplementation decreased n-6/n-3 PUFAs ratio in the brain and pancreas and blocked glucose intolerance, insulin resistance, GIIS impairment, and obesity development. The n-6/n-3 essential PUFAs in a ratio of 1.2 : 1.0 supplemented in maternal diet during pregnancy and lactation prevent metabolic dysfunction in MSG-obesity model.
Collapse
Affiliation(s)
- Josiane Morais Martin
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Rosiane Aparecida Miranda
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Luiz Felipe Barella
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Kesia Palma-Rigo
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Vander Silva Alves
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Gabriel Sergio Fabricio
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Audrei Pavanello
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | | | - Tatiane Aparecida Ribeiro
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | | | | | | | - Paulo Cezar de Freitas Mathias
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
| | - Júlio Cezar de Oliveira
- Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900 Maringá, PR, Brazil
- Instituto de Ciências da Saúde, Universidade Federal de Mato Grosso, 78557-267 Sinop, MT, Brazil
- *Júlio Cezar de Oliveira:
| |
Collapse
|
49
|
Nascimento CRB, Martinez CBDR. Daily intake of lead in Wistar rats at different ages: Biochemical, genotoxic and physiological effects. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 41:132-141. [PMID: 26694652 DOI: 10.1016/j.etap.2015.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 06/05/2023]
Abstract
The effects of the daily intake of feed containing lead (2.0 mg Pb kg(-1)) were evaluated in 45 (Pb45) and 90 (Pb90)-day-old Wistar female rats. Compared to the respective control groups, Pb45 rats consumed more feed and showed greater weight gain, but these parameters returned to control values in Pb90 rats. Higher blood glucose levels were observed in both Pb groups, whereas plasma insulin concentrations were higher in Pb45 but lower in the Pb90 group. Liver glycogen content was lower only in the Pb90 rats. There were no changes in plasma cortisol and acetylcholinesterase activity in the brain. Hematological alterations were observed only in Pb90 rats, which showed lower hemoglobin levels. In the liver, Pb45 rats showed decreased catalase and glutathione peroxidase activities and increased glutathione reductase activity, but in the Pb90 group, glutathione levels were higher. Increased hepatic lipid peroxidation and DNA damage in the lymphocytes were observed in both Pb groups. These results indicate that the daily intake of Pb for different periods results in metabolic changes and in the establishment of oxidative and genotoxic damage in female rats.
Collapse
|
50
|
De Gaetano A, Gaz C, Palumbo P, Panunzi S. A Unifying Organ Model of Pancreatic Insulin Secretion. PLoS One 2015; 10:e0142344. [PMID: 26555895 PMCID: PMC4640662 DOI: 10.1371/journal.pone.0142344] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/20/2015] [Indexed: 12/25/2022] Open
Abstract
The secretion of insulin by the pancreas has been the object of much attention over the past several decades. Insulin is known to be secreted by pancreatic β-cells in response to hyperglycemia: its blood concentrations however exhibit both high-frequency (period approx. 10 minutes) and low-frequency oscillations (period approx. 1.5 hours). Furthermore, characteristic insulin secretory response to challenge maneuvers have been described, such as frequency entrainment upon sinusoidal glycemic stimulation; substantial insulin peaks following minimal glucose administration; progressively strengthened insulin secretion response after repeated administration of the same amount of glucose; insulin and glucose characteristic curves after Intra-Venous administration of glucose boli in healthy and pre-diabetic subjects as well as in Type 2 Diabetes Mellitus. Previous modeling of β-cell physiology has been mainly directed to the intracellular chain of events giving rise to single-cell or cell-cluster hormone release oscillations, but the large size, long period and complex morphology of the diverse responses to whole-body glucose stimuli has not yet been coherently explained. Starting with the seminal work of Grodsky it was hypothesized that the population of pancreatic β-cells, possibly functionally aggregated in islets of Langerhans, could be viewed as a set of independent, similar, but not identical controllers (firing units) with distributed functional parameters. The present work shows how a single model based on a population of independent islet controllers can reproduce very closely a diverse array of actually observed experimental results, with the same set of working parameters. The model's success in reproducing a diverse array of experiments implies that, in order to understand the macroscopic behaviour of the endocrine pancreas in regulating glycemia, there is no need to hypothesize intrapancreatic pacemakers, influences between different islets of Langerhans, glycolitic-induced oscillations or β-cell sensitivity to the rate of change of glycemia.
Collapse
Affiliation(s)
- Andrea De Gaetano
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| | - Claudio Gaz
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
- Sapienza Università di Roma, Department of Computer, Control and Management Engineering (DIAG), Via Ariosto 25, 00185 Rome, Italy
| | - Pasquale Palumbo
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| | - Simona Panunzi
- CNR-IASI BioMatLab (Italian National Research Council - Institute of Analysis, Systems and Computer Science - Biomathematics Laboratory), UCSC Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|