1
|
Masloh S, Chevrel A, Culot M, Perrocheau A, Kalia YN, Frehel S, Gaussin R, Gosselet F, Huet S, Zeisser Labouebe M, Scapozza L. Enhancing Oral Delivery of Biologics: A Non-Competitive and Cross-Reactive Anti-Leptin Receptor Nanofitin Demonstrates a Gut-Crossing Capacity in an Ex Vivo Porcine Intestinal Model. Pharmaceutics 2024; 16:116. [PMID: 38258126 PMCID: PMC10820293 DOI: 10.3390/pharmaceutics16010116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Biotherapeutics exhibit high efficacy in targeted therapy, but their oral delivery is impeded by the harsh conditions of the gastrointestinal (GI) tract and limited intestinal absorption. This article presents a strategy to overcome the challenges of poor intestinal permeability by using a protein shuttle that specifically binds to an intestinal target, the leptin receptor (LepR), and exploiting its capacity to perform a receptor-mediated transport. Our proof-of-concept study focuses on the characterization and transport of robust affinity proteins, known as Nanofitins, across an ex vivo porcine intestinal model. We describe the potential to deliver biologically active molecules across the mucosa by fusing them with the Nanofitin 1-F08 targeting the LepR. This particular Nanofitin was selected for its absence of competition with leptin, its cross-reactivity with LepR from human, mouse, and pig hosts, and its shuttle capability associated with its ability to induce a receptor-mediated transport. This study paves the way for future in vivo demonstration of a safe and efficient oral-to-systemic delivery of targeted therapies.
Collapse
Affiliation(s)
- Solene Masloh
- Blood Brain Barrier Laboratory, Faculty of Science Jean Perrin, Artois University, UR 2465, Rue Jean Souvraz, 62300 Lens, France (M.C.); (F.G.)
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France (A.P.); (R.G.)
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France (A.P.); (R.G.)
| | - Maxime Culot
- Blood Brain Barrier Laboratory, Faculty of Science Jean Perrin, Artois University, UR 2465, Rue Jean Souvraz, 62300 Lens, France (M.C.); (F.G.)
| | | | - Yogeshvar N. Kalia
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Samuel Frehel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France (A.P.); (R.G.)
| | - Rémi Gaussin
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France (A.P.); (R.G.)
| | - Fabien Gosselet
- Blood Brain Barrier Laboratory, Faculty of Science Jean Perrin, Artois University, UR 2465, Rue Jean Souvraz, 62300 Lens, France (M.C.); (F.G.)
| | - Simon Huet
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France (A.P.); (R.G.)
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland (L.S.)
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
2
|
Kras K, Ropka-Molik K, Muszyński S, Arciszewski MB. Expression of Genes Encoding Selected Orexigenic and Anorexigenic Peptides and Their Receptors in the Organs of the Gastrointestinal Tract of Calves and Adult Domestic Cattle ( Bos taurus taurus). Int J Mol Sci 2023; 25:533. [PMID: 38203717 PMCID: PMC10779135 DOI: 10.3390/ijms25010533] [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: 10/02/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The regulation of food intake occurs at multiple levels, and two of the components of this process are orexigenic and anorexigenic peptides, which stimulate or inhibit appetite, respectively. The study of the function of these compounds in domestic cattle is essential for production efficiency, animal welfare, and health, as well as for economic benefits, environmental protection, and the contribution to a better understanding of physiological aspects that can be applied to other species. In this study, the real-time PCR method was utilized to determine the expression levels of GHRL, GHSR, SMIM20, GPR173, LEP, LEPR, and NUCB2 (which encode ghrelin, its receptor, phoenixin-14, its receptor, leptin, its receptor, and nesfatin-1, respectively) in the gastrointestinal tract (GIT) of Polish Holstein-Friesian breed cattle. In all analyzed GIT segments, mRNA for all the genes was present in both age groups, confirming their significance in these tissues. Gene expression levels varied distinctly across different GIT segments and between young and mature subjects. The differences between calves and adults were particularly pronounced in areas such as the forestomachs, ileum, and jejunum, indicating potential changes in peptides regulating food intake based on the developmental phase. In mature individuals, the forestomachs predominantly displayed an increase in GHRL expression, while the intestines had elevated levels of GHSR, GPR173, LEP, and NUCB2. In contrast, the forestomachs in calves showed upregulated expressions of LEP, LEPR, and NUCB2, highlighting the potential importance of peptides from these genes in bovine forestomach development.
Collapse
Affiliation(s)
- Katarzyna Kras
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12 St., 20-950 Lublin, Poland;
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Krakowska 1 St., 32-083 Balice, Poland;
| | - Siemowit Muszyński
- Department of Biophysics, Faculty of Environmental Biology, University of Life Sciences in Lublin, 13 Akademicka St., 20-950 Lublin, Poland;
| | - Marcin B. Arciszewski
- Department of Animal Anatomy and Histology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka 12 St., 20-950 Lublin, Poland;
| |
Collapse
|
3
|
Effect of Adropin on Pancreas Exocrine Function in a Rat Model: A Preliminary Study. Animals (Basel) 2022; 12:ani12192547. [PMID: 36230288 PMCID: PMC9558541 DOI: 10.3390/ani12192547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
The aim was to investigate the potential effect of adropin (ADR) on pancreatic−biliary juice (PBJ) secretion (volume, protein content, trypsin activity) in a rat model. The animals were divided into control and five experimental groups: adropin, CCK-8 (CCK-8 stimulation), capsaicin (capsaicin deactivation of afferents), vagotomy (vagotomy procedure), and vagal stimulation (vagal nerve stimulation). The experiment consisted of four phases, during which vehicle (0.9% NaCl) and three ADR boluses (5, 10, and 20 µg/kg BW) were administered i.v. every 30 min. PBJ samples were collected from each rat at 15 min intervals after boluses. Exogenous ADR failed to affect the pancreatic responses after vagotomy and the capsaicin pretreatment and reduced the PBJ volume, protein outputs, and trypsin activity in the adropin, CCK-8, and vagal stimulation groups in a dose-dependent manner. In all these groups, volume of PBJ was reduced only by the highest dose of ADR (p < 0.001 for adropin group and p < 0.01 for CCK-8 and vagal stimulation groups), and the protein outputs were reduced by the administration of ADR 10 µg/kg BW (adropin and CCK-8 groups, p < 0.01 in both cases) and 20 µg/kg BW (p < 0.001 for adropin and CCK-8 groups, p < 0.01 for vagal stimulation group). The 10 µg/kg BW dose of ADR reduced the trypsin output in the CCK-8 group (p < 0.01), and the highest ADR dose reduced the trypsin output in the CCK-8 (p < 0.001) and vagal stimulation (p < 0.01) groups. In conclusion, adropin in the analyzed doses exhibits the negative feedback pathway. This mechanism seems to participate in the regulation of pancreatic juice secretion via an indirect vagal mechanism.
Collapse
|
4
|
Ghanem M, Lewis GF, Xiao C. Recent advances in cytoplasmic lipid droplet metabolism in intestinal enterocyte. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159197. [PMID: 35820577 DOI: 10.1016/j.bbalip.2022.159197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Processing of dietary fats in the intestine is a highly regulated process that influences whole-body energy homeostasis and multiple physiological functions. Dysregulated lipid handling in the intestine leads to dyslipidemia and atherosclerotic cardiovascular disease. In intestinal enterocytes, lipids are incorporated into lipoproteins and cytoplasmic lipid droplets (CLDs). Lipoprotein synthesis and CLD metabolism are inter-connected pathways with multiple points of regulation. This review aims to highlight recent advances in the regulatory mechanisms of lipid processing in the enterocyte, with particular focus on CLDs. In-depth understanding of the regulation of lipid metabolism in the enterocyte may help identify therapeutic targets for the treatment and prevention of metabolic disorders.
Collapse
Affiliation(s)
- Murooj Ghanem
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Gary F Lewis
- Departments of Medicine and Physiology, University of Toronto, and University Health Network, Toronto, ON, Canada
| | - Changting Xiao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
5
|
Jacome-Sosa M, Hu Q, Manrique-Acevedo CM, Phair RD, Parks EJ. Human intestinal lipid storage through sequential meals reveals faster dinner appearance is associated with hyperlipidemia. JCI Insight 2021; 6:e148378. [PMID: 34369385 PMCID: PMC8489663 DOI: 10.1172/jci.insight.148378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Background It is increasingly recognized that intestinal cells can store lipids after a meal, yet the effect of this phenomenon on lipid absorption patterns in insulin resistance remains unknown. Methods The kinetics of meal fat appearance were measured in insulin-sensitive (IS, n = 8) and insulin-resistant (IR, n = 8) subjects after sequential, isotopically labeled lunch and dinner meals. Plasma dynamics on triacylglycerol-rich (TAG-rich) lipoproteins and plasma hormones were analyzed using a nonlinear, non–steady state kinetic model. Results At the onset of dinner, IS subjects showed an abrupt plasma appearance of lunch lipid consistent with the “second-meal effect,” followed by slower appearance of dinner fat in plasma, resulting in reduced accumulation of dinner TAG of 48% compared with lunch. By contrast, IR subjects exhibited faster meal TAG appearance rates after both lunch and dinner. This effect of lower enterocyte storage between meals was associated with greater nocturnal and next-morning hyperlipidemia. The biochemical data and the kinetic analysis of second-meal effect dynamics are consistent with rapid secretion of stored TAG bypassing lipolysis and resynthesis. In addition, the data are consistent with a role for the diurnal pattern of plasma leptin in regulating the processing of dietary lipid. Conclusion These data support the concept that intestinal lipid storage may be physiologically beneficial in IS subjects. Trial registration ClinicalTrials.gov NCT02020343. Funding This study was supported by a grant from the American Diabetes Association (grant 1-13-TS-12).
Collapse
Affiliation(s)
| | - Qiong Hu
- Department of Nutrition and Exercise Physiology and
| | | | - Robert D Phair
- Integrative Bioinformatics, Inc., Mountain View, California, USA
| | - Elizabeth J Parks
- Department of Nutrition and Exercise Physiology and.,Division of Gastroenterology and Hepatology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
6
|
A centric view of JAK/STAT5 in intestinal homeostasis, infection, and inflammation. Cytokine 2021; 139:155392. [PMID: 33482575 PMCID: PMC8276772 DOI: 10.1016/j.cyto.2020.155392] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Cytokines, growth factors or hormones take action through the JAK/STAT5 signaling pathway, which plays a critical role in regulating the intestinal response to infection and inflammation. However, the way in which STAT5 regulates intestinal epithelial compartment is largely ignored due to the lack of genetic tools for proper exploration and because the two STAT5 transcription factors (STAT5A and STAT5B) have some redundant but also distinct functions. In this review article, by focusing on STAT5 functions in the intestinal undifferentiated and differentiated epithelia, we discuss major advances of the growth factor/cytokine-JAK/STAT5 research in view of intestinal mucosal inflammation and immunity. We highlight the gap in the research of the intestinal STAT5 signaling to anticipate the gastrointestinal explorative insights. Furthermore, we address the critical questions to illuminate how STAT5 signaling influences intestinal epithelial cell differentiation and stem cell regeneration during homeostasis and injury. Overall, our article provides a centric view of the relevance of the relationship between chronic inflammatory diseases and JAK/STAT5 pathway and it also gives an example of how chronic infection and inflammation pirate STAT5 signaling to worsen intestinal injuries. Importantly, our review suggests how to protect a wound healing from gastrointestinal diseases by modulating intestinal STAT5.
Collapse
|
7
|
Alterations in Small Intestine and Liver Morphology, Immunolocalization of Leptin, Ghrelin and Nesfatin-1 as Well as Immunoexpression of Tight Junction Proteins in Intestinal Mucosa after Gastrectomy in Rat Model. J Clin Med 2021; 10:jcm10020272. [PMID: 33450994 PMCID: PMC7828391 DOI: 10.3390/jcm10020272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/01/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
The stomach is responsible for the processing of nutrients as well as for the secretion of various hormones which are involved in many activities throughout the gastrointestinal tract. Experimental adult male Wistar rats (n = 6) underwent a modified gastrectomy, while control rats (n = 6) were sham-operated. After six weeks, changes in small intestine (including histomorphometrical parameters of the enteric nervous plexuses) and liver morphology, immunolocalization of leptin, ghrelin and nesfatin-1 as well as proteins forming adherens and tight junctions (E-cadherin, zonula occludens-1, occludin, marvelD3) in intestinal mucosa were evaluated. A number of effects on small intestine morphology, enteric nervous system ganglia, hormones and proteins expression were found, showing intestinal enteroplasticity and neuroplasticity associated with changes in gastrointestinal tract condition. The functional changes in intestinal mucosa and the enteric nervous system could be responsible for the altered intestinal barrier and hormonal responses following gastrectomy. The results suggest that more complicated regulatory mechanisms than that of compensatory mucosal hypertrophy alone are involved.
Collapse
|
8
|
Merigo F, Brandolese A, Facchin S, Boschi F, Di Chio M, Savarino E, D'Incà R, Sturniolo GC, Sbarbati A. Immunolocalization of leptin and leptin receptor in colorectal mucosa of ulcerative colitis, Crohn's disease and control subjects with no inflammatory bowel disease. Cell Tissue Res 2020; 383:1103-1122. [PMID: 33159578 PMCID: PMC7960629 DOI: 10.1007/s00441-020-03297-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/14/2020] [Indexed: 01/04/2023]
Abstract
The expression of leptin and leptin receptor (Ob-R) has been partially elucidated in colon of patients with inflammatory bowel diseases (IBDs), even though leptin is involved in angiogenesis and inflammation. We previously reported overexpression of GLUT5 fructose transporter, in aberrant clusters of lymphatic vessels in lamina propria of IBD and controls. Here, we examine leptin and Ob-R expression in the same biopsies. Specimens were obtained from patients with ulcerative colitis (UC), Crohn’s disease (CD) and controls who underwent screening for colorectal cancer, follow-up after polypectomy or with a history of lower gastrointestinal symptoms. Immunohistochemistry revealed leptin in apical and basolateral membranes of short epithelial portions, Ob-R on the apical pole of epithelial cells. Leptin and Ob-R were also identified in structures and cells scattered in the lamina propria. In UC, a significant correlation between leptin and Ob-R in the lamina propria was found in all inflamed samples, beyond non-inflamed samples of the proximal tract, while in CD, it was found in inflamed distal samples. Most of the leptin and Ob-R positive areas in the lamina propria were also GLUT5 immunoreactive in inflamed and non-inflamed mucosa. A significant correlation of leptin or Ob-R expression with GLUT5 was observed in the inflamed distal samples from UC. Our findings suggest that there are different sites of leptin and Ob-R expression in large intestine and those in lamina propria do not reflect the status of mucosal inflammation. The co-localization of leptin and/or Ob-R with GLUT5 may indicate concomitance effects in colorectal lamina propria areas.
Collapse
Affiliation(s)
- Flavia Merigo
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37134, Verona, Italy.
| | - Alessandro Brandolese
- Department of Medicine, Gastroenterology Section, University of Verona, 37134, Verona, Italy
| | - Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, 35128, Padua, Italy
| | - Federico Boschi
- Department of Computer Science, University of Verona, 37134, Verona, Italy
| | - Marzia Di Chio
- Department of Diagnostic and Public Health, University of Verona, 37134, Verona, Italy
| | - Edoardo Savarino
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, 35128, Padua, Italy
| | - Renata D'Incà
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, 35128, Padua, Italy
| | - Giacomo Carlo Sturniolo
- Department of Surgery, Oncology and Gastroenterology, Gastroenterology Section, University Hospital of Padua, 35128, Padua, Italy
| | - Andrea Sbarbati
- Department of Neuroscience, Biomedicine and Movement, Human Anatomy and Histology Section, University of Verona, 37134, Verona, Italy
| |
Collapse
|
9
|
Inagaki-Ohara K. Gastric Leptin and Tumorigenesis: Beyond Obesity. Int J Mol Sci 2019; 20:ijms20112622. [PMID: 31141984 PMCID: PMC6600422 DOI: 10.3390/ijms20112622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022] Open
Abstract
Leptin, an adipocyte-derived hormone and its receptor (ObR) expressed in the hypothalamus are well known as an essential regulator of appetite and energy expenditure. Obesity induces abundant leptin production, however, reduced sensitivity to leptin leads to the development of metabolic disorders, so called leptin resistance. The stomach has been identified as an organ that simultaneously expresses leptin and ObR. Accumulating evidence has shown gastric leptin to perform diverse functions, such as those in nutrient absorption and carcinogenesis in the gastrointestinal system, independent of its well-known role in appetite regulation and obesity. Overexpression of leptin and phosphorylated ObR is implicated in gastric cancer in humans and in murine model, and diet-induced obesity causes precancerous lesions in the stomach in mice. While the underlying pathomechanisms remain unclear, leptin signaling can affect gastric mucosal milieu. In this review, we focus on the significant role of the gastric leptin signaling in neoplasia and tumorigenesis in stomach in the context of hereditary and diet-induced obesity.
Collapse
Affiliation(s)
- Kyoko Inagaki-Ohara
- Division of Host Defense, Department of Life Sciences, Faculty of Life and Environmental Sciences, Prefectural University of Hiroshima, 5562 Nanatsuka, Shobara, Hiroshima 727-0023, Japan.
| |
Collapse
|
10
|
Alti D, Sambamurthy C, Kalangi SK. Emergence of Leptin in Infection and Immunity: Scope and Challenges in Vaccines Formulation. Front Cell Infect Microbiol 2018; 8:147. [PMID: 29868503 PMCID: PMC5954041 DOI: 10.3389/fcimb.2018.00147] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 04/20/2018] [Indexed: 01/01/2023] Open
Abstract
Deficiency of leptin (ob/ob) and/or desensitization of leptin signaling (db/db) and elevated expression of suppressor of cytokine signaling-3 (SOCS3) reported in obesity are also reported in a variety of pathologies including hypertriglyceridemia, insulin resistance, and malnutrition as the risk factors in host defense system. Viral infections cause the elevated SOCS3 expression, which inhibits leptin signaling. It results in immunosuppression by T-regulatory cells (Tregs). The host immunity becomes incompetent to manage pathogens' attack and invasion, which results in the accelerated infections and diminished vaccine-specific antibody response. Leptin was successfully used as mucosal vaccine adjuvant against Rhodococcus equi. Leptin induced the antibody response to Helicobacter pylori vaccination in mice. An integral leptin signaling in mucosal gut epithelial cells offered resistance against Clostridium difficile and Entameoba histolytica infections. We present in this review, the intervention of leptin in lethal diseases caused by microbial infections and propose the possible scope and challenges of leptin as an adjuvant tool in the development of effective vaccines.
Collapse
Affiliation(s)
- Dayakar Alti
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| | | | - Suresh K Kalangi
- School of Life Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
11
|
Huang LL, Yang F, Xiong F. [Association of leptin, adiponectin, and ghrelin in breast milk with the growth of infants with exclusive breastfeeding]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:91-96. [PMID: 29429454 PMCID: PMC7389239 DOI: 10.7499/j.issn.1008-8830.2018.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/12/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To investigate the association of leptin, adiponectin, and ghrelin in breast milk with the weight growth velocity of infants with exclusive breastfeeding. METHODS A total of 67 full-term singleton infants who received regular child care and exclusive breastfeeding and their mothers were enrolled. The nutritional status was evaluated based on the measurements of body weight and body length (underweight, growth retardation, emaciation, overweight, and obesity). Z score was used to calculate growth velocity, and according to the ΔZ score, the infants were divided into poor growth group, low growth velocity group, and normal growth velocity group. Mature breast milk samples were collected from their mothers, and ELISA was used to measure the levels of leptin, adiponectin, and ghrelin. RESULTS The emaciation group had a significantly lower level of leptin in breast milk than the non-emaciation group (P<0.05), and the overweight/obesity group had a significantly lower level of adiponectin than the non-overweight/obesity group (P<0.05). The correlation analysis showed that the level of ghrelin in breast milk was positively correlated with Z score of current body weight and ΔZ score compared with birth weight (rs=0.280 and 0.290 respectively; P<0.05). The regression analysis showed that the level of ghrelin in breast milk was an important influencing factor for the Z score of body weight (β=0.161, P<0.05). CONCLUSIONS Various active constituents in breast milk, including leptin, adiponectin, and ghrelin, may regulate the growth and development of infants to a certain degree, but long-term studies and observation are needed to investigate their association with offspring growth and development and the health-promoting effect of breast milk on offspring.
Collapse
Affiliation(s)
- Li-Li Huang
- Department of Pediatrics, West China Second University Hospital, Sichuan University/Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China.
| | | | | |
Collapse
|
12
|
Yahya RS, Awad SI, Kizilbash N, El-Baz HA, Atia G. Enteric parasites can disturb leptin and adiponectin levels in children. Arch Med Sci 2018; 14:101-106. [PMID: 29379539 PMCID: PMC5778414 DOI: 10.5114/aoms.2016.60707] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 08/01/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Infection by intestinal parasites in childhood may be the main cause of many health-related problems in developed countries such as anemia, anorexia, loss of appetite, retarded growth and development. The aim of the present study was to assess the effect of different intestinal parasites on white adipose tissue hormones. MATERIAL AND METHODS Eighty-one children infected by different parasites and 35 apparently healthy children were enrolled in this study. All patients and controls were subjected to clinical examination, measurement of body mass index (BMI) and laboratory examination. RESULTS For BMI percentiles, there was a significant increase in serum leptin level (p = 0.042) and a significant decrease in serum adiponectin level (p = 0.039) in uninfected children, whereas there were no significant changes in the infected group (p = 0.068 and 0.082 respectively). A significant increase in leptin and decrease in adiponectin levels were observed for E. histolytica, Strongyloides and E. histolytica and Giardia infections compared to the control group (p = 0.047, 0.035 and 0.019 for leptin, and p = 0.025, 0.038 and 0.041 for adiponectin, respectively). CONCLUSIONS The infection by some intestinal parasites may deregulate the secretion of leptin and adiponectin and also affect the absorption of some nutrients which can disturb the BMI and cause anorexia.
Collapse
Affiliation(s)
- Raida S Yahya
- Department of Laboratory Medicine, Children's' Hospital, Faculty of Medicine, Mansoura University, Egypt
| | - Soha I Awad
- Department of Parasitology, Faculty of Medicine, Mansoura, Egypt
- Department of Medical Parasitology, Faculty of Medicine and Applied Medical Sciences, Northern Border University, Saudi Arabia
| | - Nadeem Kizilbash
- Department of Biochemistry, Faculty of Medicine and Applied Medical Sciences, Northern Border University, Arar, Saudi Arabia
| | - Hatim A El-Baz
- Department of Biochemistry, National Research Centre, Cairo, Egypt
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University (North Jeddah Branch), Saudi Arabia
| | - Gehan Atia
- Department of Pediatrics, Children Hospital, Faculty of Medicine, Mansoura, Egypt
| |
Collapse
|
13
|
Poljaroen J, Tinikul Y, Tinikul R, Anurucpreeda P, Sobhon P. Leptin-like immunoreactivity in the central nervous system, digestive organs, and gonads of the giant freshwater prawn, Macrobrachium rosenbergii. Acta Histochem 2017. [PMID: 28624121 DOI: 10.1016/j.acthis.2017.06.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Leptin, a highly conserved adipocyte-derived hormone, plays important roles in a variety of physiological processes, including the control of fat storage and metabolic status which are linked to food intake, energy homeostasis, and reproduction in all vertebrates. In the present study, we hypothesize that leptin is also present in various organs of the fresh water prawns, Macrobrachium rosenbergii. The existence and distribution of a leptin-like peptide in prawn tissues were verified by using Western blotting (WB) and immunohistochemical detection (ID) using primary antibody against human leptin. With WB, a leptin-like peptide, having a molecular weight of 15kDa, was detected in the brain, thoracic ganglia, abdominal ganglia, parts of the gastro-intestinal tract, hepatopancreas, adipocytes and gonads. By ID, leptin immunoreactivity (leptin-ir) was detected in the brain, thoracic ganglia and intersegmental commissural nerve fibers of abdominal ganglia. In the gastrointestinal tract, there was intense leptin-ir in the apical part of the epithelial cells of the cardiac and pyloric parts of the stomach. In the midgut and hindgut, the leptin-ir was detected in epithelial cells and basal cells located near the basal lamina of the epithelium. In addition, there was leptin-ir in the Restzellen cells in the hepatopancreas which produce digestive enzymes. In the ovary, the strong intensity of a leptin-ir was detected in the cytoplasm of middle to late stage oocytes, whereas no positive staining was detected in follicular cells. An intense leptin-ir was detected in spermatocytes and sustentacular cells in the seminiferous tubules in the testes of small and orange claw males. Taken together, the detection of the leptin-ir in several organs implicates the existence of a leptin-like peptide in various organs of the freshwater prawn; and like in vertebrates this peptide may be an important hormonal factor in controlling feeding and reproductive process.
Collapse
|
14
|
Abstract
This review discusses the regulation of the intestinal and hypothalamic apolipoprotein A-IV (apo A-IV) gene and protein expression. Apo A-IV is a glycoprotein secreted together with triglyceride-rich lipoproteins by the small intestine. Intestinal apo A-IV synthesis is stimulated by fat absorption, probably mediated by chylomicron formation. This stimulation of intestinal apo A-IV synthesis is attenuated by intravenous leptin infusion. Chronic ingestion of a high-fat diet blunts the intestinal apo A-IV in response to dietary lipid. Intestinal apo A-IV synthesis is also stimulated by members of the pancreatic polypeptide family, including peptide YY (PYY), neuropeptide Y (NPY), and pancreatic polypeptide (PP). Recently, apo A-IV was demonstrated to be present in the hypothalamus as well. Hypothalamic apo A-IV level was reduced by food deprivation and restored by lipid feeding. Intracerebroventricular administration of apo A-IV antiserum stimulated feeding and decreased the hypothalamic apo A-IV mRNA level, implying that feeding is intimately regulated by endogenous hypothalamic apo A-IV. Central administration of NPY significantly increased hypothalamic apo A-IV mRNA levels in a dose-dependent manner.
Collapse
Affiliation(s)
- Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical College, Cincinnati, Ohio 45267-0529, USA
| | | | | |
Collapse
|
15
|
D'Aquila T, Hung YH, Carreiro A, Buhman KK. Recent discoveries on absorption of dietary fat: Presence, synthesis, and metabolism of cytoplasmic lipid droplets within enterocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:730-47. [PMID: 27108063 DOI: 10.1016/j.bbalip.2016.04.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/16/2016] [Accepted: 04/16/2016] [Indexed: 02/07/2023]
Abstract
Dietary fat provides essential nutrients, contributes to energy balance, and regulates blood lipid concentrations. These functions are important to health, but can also become dysregulated and contribute to diseases such as obesity, diabetes, cardiovascular disease, and cancer. Within enterocytes, the digestive products of dietary fat are re-synthesized into triacylglycerol, which is either secreted on chylomicrons or stored within cytoplasmic lipid droplets (CLDs). CLDs were originally thought to be inert stores of neutral lipids, but are now recognized as dynamic organelles that function in multiple cellular processes in addition to lipid metabolism. This review will highlight recent discoveries related to dietary fat absorption with an emphasis on the presence, synthesis, and metabolism of CLDs within this process.
Collapse
Affiliation(s)
- Theresa D'Aquila
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Yu-Han Hung
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Alicia Carreiro
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA
| | - Kimberly K Buhman
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
16
|
Rieg JAD, Chirasani VR, Koepsell H, Senapati S, Mahata SK, Rieg T. Regulation of intestinal SGLT1 by catestatin in hyperleptinemic type 2 diabetic mice. J Transl Med 2016; 96:98-111. [PMID: 26552046 PMCID: PMC4695279 DOI: 10.1038/labinvest.2015.129] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/10/2015] [Accepted: 09/24/2015] [Indexed: 01/23/2023] Open
Abstract
The small intestine is the major site for nutrient absorption that is critical in maintenance of euglycemia. Leptin, a key hormone involved in energy homeostasis, directly affects nutrient transport across the intestinal epithelium. Catestatin (CST), a 21-amino acid peptide derived from proprotein chromogranin A, has been shown to modulate leptin signaling. Therefore, we reasoned that leptin and CST could modulate intestinal Na(+)-glucose transporter 1 (SGLT1) expression in the context of obesity and diabetes. We found that hyperleptinemic db/db mice exhibit increased mucosal mass, associated with an enhanced proliferative response and decreased apoptosis in intestinal crypts, a finding absent in leptin-deficient ob/ob mice. Intestinal SGLT1 abundance was significantly decreased in hyperleptinemic but not leptin-deficient mice, indicating leptin regulation of SGLT1 expression. Phlorizin, a SGLT1/2 inhibitor, was without effect in an oral glucose tolerance test in db/db mice. The alterations in architecture and SGLT1 abundance were not accompanied by changes in the localization of intestinal alkaline phosphatase, indicating intact differentiation. Treatment of db/db mice with CST restored intestinal SGLT1 abundance and intestinal turnover, suggesting a cross-talk between leptin and CST, without affecting plasma leptin levels. Consistent with this hypothesis, we identified structural homology between CST and the AB-loop of leptin and protein-protein docking revealed binding of CST and leptin with the Ig-like binding site-III of the leptin receptor. In summary, downregulation of SGLT1 in an obese type 2 diabetic mouse model with hyperleptinemia is presumably mediated via the short form of the leptin receptor and reduces overt hyperglycemia.
Collapse
Affiliation(s)
- Jessica A. Dominguez Rieg
- Department of Basic Sciences, Bastyr University California, San Diego, CA, USA,VA San Diego Healthcare System, San Diego, California; CA, USA
| | | | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Sanjib Senapati
- Department of Biotechnology, Institute of Technology Madras, Chennai, India
| | - Sushil K. Mahata
- VA San Diego Healthcare System, San Diego, California; CA, USA,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timo Rieg
- VA San Diego Healthcare System, San Diego, California; CA, USA,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Allensworth-James ML, Odle A, Haney A, Childs G. Sex Differences in Somatotrope Dependency on Leptin Receptors in Young Mice: Ablation of LEPR Causes Severe Growth Hormone Deficiency and Abdominal Obesity in Males. Endocrinology 2015; 156:3253-64. [PMID: 26168341 PMCID: PMC4541611 DOI: 10.1210/en.2015-1198] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leptin receptor (LEPR) signaling controls appetite and energy expenditure. Somatotrope-specific deletion of the LEPRb signaling isoform causes GH deficiency and obesity. The present study selectively ablated Lepr exon 1 in somatotropes, which removes the signal peptide, causing the loss of all isoforms of LEPR. Excision of Lepr exon 1 was restricted to the pituitary, and mutant somatotropes failed to respond to leptin. Young (2-3 mo) males showed a severe 84% reduction in serum GH levels and more than 60% reduction in immunolabeled GH cells compared with 41%-42% reductions in GH and GH cells in mutant females. Mutant males (35 d) and females (45 d) weighed less than controls and males had lower lean body mass. Image analysis of adipose tissue by magnetic resonance imaging showed that young males had a 2-fold increase in abdominal fat mass and increased adipose tissue density. Young females had only an overall increase in adipose tissue. Both males and females showed lower energy expenditure and higher respiratory quotient, indicating preferential carbohydrate burning. Young mutant males slept less and were more restless during the dark phase, whereas the opposite was true of females. The effects of a Cre-bearing sire on his non-Cre-recombinase bearing progeny are seen by increased respiratory quotient and reduced litter sizes. These studies elucidate clear sex differences in the extent to which somatotropes are dependent on all isoforms of LEPR. These results, which were not seen with the ablation of Lepr exon 17, highlight the severe consequences of ablation of LEPR in male somatotropes.
Collapse
Affiliation(s)
- Melody L Allensworth-James
- Department of Neurobiology and Developmental Sciences, College of Medicine University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212
| | - Angela Odle
- Department of Neurobiology and Developmental Sciences, College of Medicine University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212
| | - Anessa Haney
- Department of Neurobiology and Developmental Sciences, College of Medicine University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212
| | - Gwen Childs
- Department of Neurobiology and Developmental Sciences, College of Medicine University of Arkansas for Medical Sciences, Little Rock, Arkansas 72212
| |
Collapse
|
18
|
Rault JL, Ferrari J, Pluske JR, Dunshea FR. Neonatal oxytocin administration and supplemental milk ameliorate the weaning transition and alter hormonal expression in the gastrointestinal tract in pigs. Domest Anim Endocrinol 2015; 51:19-26. [PMID: 25481273 DOI: 10.1016/j.domaniend.2014.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 12/12/2022]
Abstract
The aim of this study was to investigate the influences of milk supplementation during lactation, over 1 wk after weaning, and oxytocin administration for the first 14 d of life on the pigs' response to weaning. Pigs from 20 litters were allocated to each of these 3 treatments in a randomized factorial design. Oxytocin was administered subcutaneously daily from 0 to 14 d of age at a rate of 10 I.U. per kg. The milk supplement consisted of a mixture of 25% skim milk powder offered either during lactation between 10 and 20 d of age or for the first week after weaning as a transitional diet along with dry pellets. Pigs were weaned at 21 d of age. Growth rate was measured from birth to slaughter at 140 d of age and feed intake of supplemental milk or feed from 10 to 56 d of age. Organ weights (heart, liver, stomach, and kidneys) and the gene expression of ghrelin, leptin, and glucagon-like peptides (glucagon-like peptide-1 and glucagon-like peptide-2) were measured in the stomach, ileum, and duodenum at 10, 21, and 28 d of age. Milk supplementation after weaning resulted in immediate feed intake and partially alleviated the depression in growth rate over the first 7 d postweaning (P < 0.001), but milk supplementation during lactation had no effects (P > 0.1). However, effects were only transient and disappeared once the milk liquid diet was removed. Neonatal oxytocin administration reduced weight loss over the first 2 d after weaning (P = 0.03), without affecting feed intake (P > 0.1), hence possibly reducing weaning stress. Seven days after weaning, oxytocin-treated pigs had greater stomach ghrelin and leptin expression (both P = 0.02), and pigs supplemented with milk after weaning had greater stomach leptin and glucagon-like peptide-2 expression (P = 0.02 and P = 0.05, respectively). Hence, neonatal oxytocin administration or postweaning milk supplementation are both effective means of enhancing gastric leptin expression and reducing weight loss at weaning, likely improving gut health during this critical period.
Collapse
Affiliation(s)
- J-L Rault
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Victoria 3010, Australia
| | - J Ferrari
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Victoria 3010, Australia; School of Veterinary and Life Sciences, Murdoch University, Western Austrailia 6150, Australia
| | - J R Pluske
- School of Veterinary and Life Sciences, Murdoch University, Western Austrailia 6150, Australia
| | - F R Dunshea
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
19
|
Kohan AB, Wang F, Lo CM, Liu M, Tso P. ApoA-IV: current and emerging roles in intestinal lipid metabolism, glucose homeostasis, and satiety. Am J Physiol Gastrointest Liver Physiol 2015; 308:G472-81. [PMID: 25591862 PMCID: PMC4360046 DOI: 10.1152/ajpgi.00098.2014] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Apolipoprotein A-IV (apoA-IV) is secreted by the small intestine on chylomicrons into intestinal lymph in response to fat absorption. Many physiological functions have been ascribed to apoA-IV, including a role in chylomicron assembly and lipid metabolism, a mediator of reverse-cholesterol transport, an acute satiety factor, a regulator of gastric function, and, finally, a modulator of blood glucose homeostasis. The purpose of this review is to update our current view of intestinal apoA-IV synthesis and secretion and the physiological roles of apoA-IV in lipid metabolism and energy homeostasis, and to underscore the potential for intestinal apoA-IV to serve as a therapeutic target for the treatment of diabetes and obesity-related disease.
Collapse
Affiliation(s)
- Alison B. Kohan
- 2Department of Nutritional Sciences, University of Connecticut, Storrs, Connecticut
| | - Fei Wang
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Chun-Min Lo
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Min Liu
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Patrick Tso
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| |
Collapse
|
20
|
Wang F, Kohan AB, Lo CM, Liu M, Howles P, Tso P. Apolipoprotein A-IV: a protein intimately involved in metabolism. J Lipid Res 2015; 56:1403-18. [PMID: 25640749 DOI: 10.1194/jlr.r052753] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Indexed: 01/07/2023] Open
Abstract
The purpose of this review is to summarize our current understanding of the physiological roles of apoA-IV in metabolism, and to underscore the potential for apoA-IV to be a focus for new therapies aimed at the treatment of diabetes and obesity-related disorders. ApoA-IV is primarily synthesized by the small intestine, attached to chylomicrons by enterocytes, and secreted into intestinal lymph during fat absorption. In circulation, apoA-IV is associated with HDL and chylomicron remnants, but a large portion is lipoprotein free. Due to its anti-oxidative and anti-inflammatory properties, and because it can mediate reverse-cholesterol transport, proposed functions of circulating apoA-IV have been related to protection from cardiovascular disease. This review, however, focuses primarily on several properties of apoA-IV that impact other metabolic functions related to food intake, obesity, and diabetes. In addition to participating in triglyceride absorption, apoA-IV can act as an acute satiation factor through both peripheral and central routes of action. It also modulates glucose homeostasis through incretin-like effects on insulin secretion, and by moderating hepatic glucose production. While apoA-IV receptors remain to be conclusively identified, the latter modes of action suggest that this protein holds therapeutic promise for treating metabolic disease.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237
| | - Alison B Kohan
- Department of Nutritional Sciences, University of Connecticut Advanced Technology Laboratory, Storrs, CT 06269
| | - Chun-Min Lo
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237
| | - Min Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237
| | - Philip Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45237
| |
Collapse
|
21
|
Sáinz N, Barrenetxe J, Moreno-Aliaga MJ, Martínez JA. Leptin resistance and diet-induced obesity: central and peripheral actions of leptin. Metabolism 2015; 64:35-46. [PMID: 25497342 DOI: 10.1016/j.metabol.2014.10.015] [Citation(s) in RCA: 315] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 10/15/2014] [Accepted: 10/20/2014] [Indexed: 12/15/2022]
Abstract
Obesity is a chronic disease that represents one of the most serious global health burdens associated to an excess of body fat resulting from an imbalance between energy intake and expenditure, which is regulated by environmental and genetic interactions. The adipose-derived hormone leptin acts via a specific receptor in the brain to regulate energy balance and body weight, although this protein can also elicit a myriad of actions in peripheral tissues. Obese individuals, rather than be leptin deficient, have in most cases, high levels of circulating leptin. The failure of these high levels to control body weight suggests the presence of a resistance process to the hormone that could be partly responsible of disturbances on body weight regulation. Furthermore, leptin resistance can impair physiological peripheral functions of leptin such as lipid and carbohydrate metabolism and nutrient intestinal utilization. The present document summarizes those findings regarding leptin resistance development and the role of this hormone in the development and maintenance of an obese state. Thus, we focused on the effect of the impaired leptin action on adipose tissue, liver, skeletal muscle and intestinal function and the accompanying relationships with diet-induced obesity. The involvement of some inflammatory mediators implicated in the development of obesity and their roles in leptin resistance development are also discussed.
Collapse
Affiliation(s)
- Neira Sáinz
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Jaione Barrenetxe
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Alfredo Martínez
- Department of Nutrition, Food Sciences and Physiology, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
22
|
El-Zein O, Usta J, El Moussawi L, Kreydiyyeh SI. Leptin inhibits the Na(+)/K(+) ATPase in Caco-2 cells via PKC and p38MAPK. Cell Signal 2014; 27:416-23. [PMID: 25499980 DOI: 10.1016/j.cellsig.2014.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 11/22/2014] [Accepted: 12/06/2014] [Indexed: 10/24/2022]
Abstract
We demonstrated previously an inhibitory effect of luminal leptin on glucose absorption in differentiated Caco-2 cells. Since this process is dependent on the Na(+) gradient established by the Na(+)/K(+)ATPase this work was undertaken to investigate if the ATPase is one of the hormone's targets. Fully differentiated Caco-2 cells were incubated with 10nM luminal leptin and the activity of the Na(+)/K(+) ATPase was assayed by measuring the amount of inorganic phosphate liberated. To elucidate the signaling pathway involved, the suspected mediators, namely PKC, p38MAPK, ERK and PI3K, were inhibited with specific pharmacological inhibitors and their implication was confirmed by determining changes in the protein expression of their active phosphorylated forms by Western blot analysis. Leptin reduced significantly the activity of the Na(+)/K(+) ATPase, by activating p38MAPK via inhibition of PKC, an upstream inhibitor of the kinase. ERK and PI3K are modulators of the pump and are not along the pathway activated by leptin but cross talk with it at the level of p38MAPK.
Collapse
Affiliation(s)
- Ola El-Zein
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | - Julnar Usta
- Department of Biochemistry & Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Layla El Moussawi
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
23
|
Khodabakhshi A, Ghayour-Mobarhan M, Rooki H, Vakili R, Hashemy SI, Mirhafez SR, Shakeri MT, Kashanifar R, Pourbafarani R, Mirzaei H, Dahri M, Mazidi M, Ferns G, Safarian M. Comparative measurement of ghrelin, leptin, adiponectin, EGF and IGF-1 in breast milk of mothers with overweight/obese and normal-weight infants. Eur J Clin Nutr 2014; 69:614-8. [DOI: 10.1038/ejcn.2014.205] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/12/2014] [Accepted: 08/22/2014] [Indexed: 11/09/2022]
|
24
|
O'Sullivan KE, Reynolds JV, O'Hanlon C, O'Sullivan JN, Lysaght J. Could signal transducer and activator of transcription 3 be a therapeutic target in obesity-related gastrointestinal malignancy? J Gastrointest Cancer 2014; 45:1-11. [PMID: 24163144 DOI: 10.1007/s12029-013-9555-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION A large body of evidence has implicated the signal transducer and activator of transcription (STAT) family and particularly the ubiquitously expressed STAT3 protein in the pathogenesis of colorectal, hepatocellular, gastric and pancreatic carcinoma. DISCUSSION Concomitantly, an increasing body of epidemiological evidence has linked obesity and its associated pro-inflammatory state with the development of gastrointestinal cancers. Visceral adipose tissue is no longer considered inert and is known to secrete a number of adipocytokines such as leptin, interleukin (IL)-6, IL-8, IL-1β and tumour necrosis factor-alpha (TNF-α) into the surrounding environment. Interestingly, these adipocytokines are strongly linked with the Janus kinase (JAK)/STAT pathway of signal transduction and there is experimental evidence linking IL-1β, IL-8 and TNF-α to JAK/STAT signaling in other tissues. The result is an up-regulation of a wide range of anti-apoptotic, pro-metastatic and pro-angiogenic genes and processes. This is particularly relevant for gastrointestinal malignancy as these factors have the potential to signal adjacent endothelial cells in a paracrine manner. CONCLUSION This review examines the potential role of the STAT3 signaling pathway in the pathogenesis of obesity-related gastrointestinal malignancy and the potential therapeutic role of STAT3 blockade given its status as a signaling hub for a number of inflammatory adipocytokines.
Collapse
Affiliation(s)
- Katie E O'Sullivan
- Department of Surgery, Institute of Molecular Medicine, St. James Hospital, Dublin 8, Ireland,
| | | | | | | | | |
Collapse
|
25
|
Guo S, Chen QP, Lin XT, Zhang F, Zhang XY. Relationship between changes in intestinal mucosal leptin/leptin receptor expression and proliferation and apoptosis of intestinal mucosal cells in patients with obstructive jaundice. Shijie Huaren Xiaohua Zazhi 2014; 22:3510-3516. [DOI: 10.11569/wcjd.v22.i23.3510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the changes in leptin (Lp)/leptin receptor (Ob-R) expression, and cell proliferation and apoptosis in the small intestinal mucosa of patients with obstructive jaundice (OJ), and to analyze their relationship.
METHODS: Thirty patients with OJ treated at our hospital from March 2012 to July 2013 were selected as the experimental group. They were further divided into a mild jaundice subgroup ( ≤ 171 μmol/L, OJ1 subgroup, n = 17) and a moderate to severe jaundice subgroup ( > 171 μmol/L, OJ2 subgroup, n = 13). Twenty-one patients without OJ were selected as negative controls. Immunohistochemistry was performed to detect he expression of Lp, Ob-R and Ki67 in the intestinal mucosa, TUNEL was used to detect the apoptosis of intestinal mucosal cells, and the intestinal villus height and crypt depth were measured by HE staining.
RESULTS: The expression of Lp and Ob-R in the small intestinal mucosa of patients with OJ was significantly lower than that in the control group (Lp: 1.63 ± 1.25, 2.48 ± 1.25, P < 0.05; Ob-R: 2.63 ± 1.27, 3.90 ± 1.00, P < 0.05). Lp/Ob-R expression was positively correlated with proliferation of intestinal epithelial cells, but negatively correlated with apoptosis.
CONCLUSION: There is a significant correlation between the expression of intestinal mucosa Lp/Ob-R and intestinal mucosal cell proliferation and apoptosis. Lp exerts positive effects in the small intestinal mucosa of patients with OJ.
Collapse
|
26
|
Tavernier A, Cavin JB, Le Gall M, Ducroc R, Denis RGP, Cluzeaud F, Guilmeau S, Sakar Y, Barbot L, Kapel N, Le Beyec J, Joly F, Chua S, Luquet S, Bado A. Intestinal deletion of leptin signaling alters activity of nutrient transporters and delayed the onset of obesity in mice. FASEB J 2014; 28:4100-10. [PMID: 24928195 DOI: 10.1096/fj.14-255158] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 05/27/2014] [Indexed: 11/11/2022]
Abstract
The importance of B-isoform of leptin receptor (LEPR-B) signaling in the hypothalamus, pancreas, or liver has been well characterized, but in the intestine, a unique site of entry for dietary nutrition into the body, it has been relatively ignored. To address this question, we characterized a mouse model deficient for LEPR-B specifically in intestinal epithelial cells (IECs). (IEC)LEPR-B-knockout (KO) and wild-type (WT) mice were generated by Cre-Lox strategy and fed a normal or high-fat diet (HFD). The analyses of the animals involved histology and immunohistochemistry of intestinal mucosa, indirect calorimetric measurements, whole-body composition, and expression and activities of nutrient transporters. (IEC)LEPR-B-KO mice exhibited a 2-fold increase in length of jejunal villi and have normal growth on a normal diet but were less susceptible (P<0.01) to HFD-induced obesity. No differences occurred in energy intake and expenditure between (IEC)LEPR-B-WT and -KO mice, but (IEC)LEPR-B-KO mice fed an HFD showed increased excreted fats (P<0.05). Activities of the Na(+)/glucose cotransporter SGLT-1 and GLUT2 were unaffected in LEPR-B-KO jejunum, while GLUT5-mediated fructose transport and PepT1-mediated peptide transport were substantially reduced (P<0.01). These data demonstrate that intestinal LEPR-B signaling is important for the onset of diet-induced obesity. They suggest that intestinal LEPR-B could be a potential per os target for prevention against obesity.
Collapse
Affiliation(s)
- Annabelle Tavernier
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Jean-Baptiste Cavin
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Maude Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Robert Ducroc
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Raphaël G P Denis
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8251, Paris, France
| | - Françoise Cluzeaud
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Sandra Guilmeau
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Yassine Sakar
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France
| | - Laurence Barbot
- Service de Coprologie Fonctionnelle, Hopital La Pitié Salpêtrière, Paris, France
| | - Nathalie Kapel
- Service de Coprologie Fonctionnelle, Hopital La Pitié Salpêtrière, Paris, France
| | - Johanne Le Beyec
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France; Université Pierre et Marie Curie, Paris, France
| | - Francisca Joly
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France; Service de Gastroenterologie et d'Assistance Nutritive, Hopital Beaujon, Clichy, France; and
| | - Streamson Chua
- Department of Medicine and Neuroscience, Albert Einstein College of Medicine, New York, New York, USA
| | - Serge Luquet
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 8251, Paris, France
| | - Andre Bado
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité Mixte de Recherche en Santé (UMRS) 1149, Unité de Formation et de Recherche (UFR) de Médecine Paris Diderot, Sorbonne Paris Cite, Paris, France;
| |
Collapse
|
27
|
Côté CD, Zadeh-Tahmasebi M, Rasmussen BA, Duca FA, Lam TKT. Hormonal signaling in the gut. J Biol Chem 2014; 289:11642-11649. [PMID: 24577102 DOI: 10.1074/jbc.o114.556068] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gut is anatomically positioned to play a critical role in the regulation of metabolic homeostasis, providing negative feedback via nutrient sensing and local hormonal signaling. Gut hormones, such as cholecystokinin (CCK) and glucagon-like peptide-1 (GLP-1), are released following a meal and act on local receptors to regulate glycemia via a neuronal gut-brain axis. Additionally, jejunal nutrient sensing and leptin action are demonstrated to suppress glucose production, and both are required for the rapid antidiabetic effect of duodenal jejunal bypass surgery. Strategies aimed at targeting local gut hormonal signaling pathways may prove to be efficacious therapeutic options to improve glucose control in diabetes.
Collapse
Affiliation(s)
- Clémence D Côté
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 1L7; Departments of Physiology, University of Toronto, Toronto, Ontario M5S 1A8
| | - Melika Zadeh-Tahmasebi
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 1L7; Departments of Physiology, University of Toronto, Toronto, Ontario M5S 1A8
| | - Brittany A Rasmussen
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 1L7; Departments of Physiology, University of Toronto, Toronto, Ontario M5S 1A8
| | - Frank A Duca
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 1L7; Departments of Medicine, University of Toronto, Toronto, Ontario M5S 1A8
| | - Tony K T Lam
- Toronto General Research Institute, University Health Network, Toronto, Ontario M5G 1L7; Departments of Physiology, University of Toronto, Toronto, Ontario M5S 1A8; Departments of Medicine, University of Toronto, Toronto, Ontario M5S 1A8; Departments of Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
28
|
Rasmussen BA, Breen DM, Duca FA, Côté CD, Zadeh-Tahmasebi M, Filippi BM, Lam TKT. Jejunal leptin-PI3K signaling lowers glucose production. Cell Metab 2014; 19:155-61. [PMID: 24361011 DOI: 10.1016/j.cmet.2013.11.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/08/2013] [Accepted: 11/11/2013] [Indexed: 10/25/2022]
Abstract
The fat-derived hormone leptin binds to its hypothalamic receptors to regulate glucose homeostasis. Leptin is also synthesized in the stomach and subsequently binds to its receptors expressed in the intestine, although the functional relevance of such activation remains largely unknown. We report here that intrajejunal leptin administration activates jejunal leptin receptors and signals through a phosphatidylinositol 3-kinase (PI3K)-dependent and signal transducer and activator of transcription 3 (STAT3)-independent signaling pathway to lower glucose production in healthy rodents. Jejunal leptin action is sufficient to lower glucose production in uncontrolled diabetic and high-fat-fed rodents and contributes to the early antidiabetic effect of duodenal-jejunal bypass surgery. These data unveil a glucoregulatory site of leptin action and suggest that enhancing leptin-PI3K signaling in the jejunum lowers plasma glucose concentrations in diabetes.
Collapse
Affiliation(s)
- Brittany A Rasmussen
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Danna M Breen
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Frank A Duca
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Clémence D Côté
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Melika Zadeh-Tahmasebi
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Beatrice M Filippi
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tony K T Lam
- Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
29
|
El-Zein O, Kreydiyyeh SI. Leptin inhibits glucose intestinal absorption via PKC, p38MAPK, PI3K and MEK/ERK. PLoS One 2013; 8:e83360. [PMID: 24340098 PMCID: PMC3858357 DOI: 10.1371/journal.pone.0083360] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/11/2013] [Indexed: 11/25/2022] Open
Abstract
The role of leptin in controlling food intake and body weight is well recognized, but whether this is achieved by modulating nutrient absorption is still a controversial issue. The aim of this work was to investigate the direct effect of luminal leptin on glucose intestinal absorption and elucidate for the first time its signaling pathway. Fully differentiated Caco-2 cells grown on transwell filters were used for glucose transport studies. Leptin caused a significant reduction in glucose absorption. Individual and simultaneous inhibition of ERK, p38MAPK, PI3K or PKC abrogated completely the inhibitory effect of leptin. Activating PKC, lead to a stimulatory effect that appeared only when ERK, p38MAPK, or PI3K was inactive. Moreover, leptin increased the phosphorylation of ERK, Akt and p38MAPK. This increase changed into a decrease when p38MAPK and PKC were inactivated individually. Inhibiting ERK maintained the leptin-induced up-regulation of p-Akt and p-p38MAPK while inhibiting PI3K reduced the level of p-ERK and p-Akt but maintained the increase in p-p38MAPK. These results suggest that leptin reduces glucose absorption by activating PKC. Although the latter modulates glucose absorption via a stimulatory and an inhibitory pathway, only the latter is involved in leptin’s action. Active PKC leads to a sequential activation of p38MAPK, PI3K and ERK which exerts an inhibitory effect on glucose absorption. The results reveal a modulatory role of leptin in nutrient absorption in addition to its known satiety inducing effect.
Collapse
Affiliation(s)
- Ola El-Zein
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | | |
Collapse
|
30
|
Kim CY, Kim KH. Curcumin prevents leptin-induced tight junction dysfunction in intestinal Caco-2 BBe cells. J Nutr Biochem 2013; 25:26-35. [PMID: 24314862 DOI: 10.1016/j.jnutbio.2013.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 08/05/2013] [Accepted: 08/30/2013] [Indexed: 12/11/2022]
Abstract
Maintaining tight junction (TJ) integrity in the intestine is critical for nutrient absorption, host defense, and host immunity. While leptin secreted from adipose tissue is associated with obesity and obesity-related intestinal inflammation, the role of luminal leptin in intestinal TJ function is elusive. Here, we examined the role of leptin in intestinal TJ function in Caco-2 BBe cells and further explored the function of curcumin (CCM) in leptin-induced TJ dysfunction. Apical leptin, but not basolateral leptin, treatment at a concentration of 100 ng/ml deteriorated TJ function in Caco-2 BBe cells. Leptin-impaired TJ alteration was resulted from induction of leptin receptor-dependent JAK2/STAT3 signaling pathway and its-related PI3K/Akt/ERK1/2 signaling pathways. Apical leptin also lowered the expression levels of genes encoding TJ-associated proteins such as zonula occludens-3, claudin-5, and occludin, and elevated expression of pro-inflammatory genes such as IL-6 and TNF-α. Leptin-impaired TJ junction in Caco-2 BBe cells was blunted by a 30-min CCM pretreatment through inhibition of leptin receptor-dependent signaling pathway, and its-associated induction of expression of genes encoding TJ-associated proteins and pro-inflammatory cytokines. Our results elucidate a novel function of luminal leptin in intestinal TJ dysfunction, and further identify CCM as an effective dietary compound that prevents leptin-impaired TJ function in intestinal cells.
Collapse
Affiliation(s)
- Choon Young Kim
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | | |
Collapse
|
31
|
The esophagitis to adenocarcinoma sequence; the role of inflammation. Cancer Lett 2013; 345:182-9. [PMID: 23994342 DOI: 10.1016/j.canlet.2013.08.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 08/08/2013] [Accepted: 08/13/2013] [Indexed: 12/19/2022]
Abstract
Esophageal adenocarcinoma (EAC) is the eighth most common cancer worldwide, and approximately 15% of patients survive 5years. Reflux disease (GERD) and Barrett's esophagus (BE) are major risk factors for the development of EAC, and epidemiologic studies highlight a strong association with obesity. The immune, inflammatory and intracellular signaling changes resulting from chronic inflammation of the esophageal squamous epithelium are increasingly well characterized. In GERD and Barrett's, an essential role for T-cells in the initiation of inflammation in the esophagus has been identified, and a balance between T-cell responses and phenotype may play an important role in disease progression. Obesity is a chronic low-grade inflammatory state, fueled by adipose tissue derived- inflammatory mediators such as IL-6, TNF-α and leptin, representing a novel area for targeted research. Additionally, reactive oxygen species (ROS) and receptor tyrosine kinase (RTK) activation may drive progression from esophagitis to EAC, and downstream signaling pathways employed by these molecules may be important. This review will explain the diverse range of mechanisms potentially driving and maintaining inflammation within the esophagus and explore both existing and future therapeutic strategies targeting the process.
Collapse
|
32
|
Bosier B, Bellocchio L, Metna-Laurent M, Soria-Gomez E, Matias I, Hebert-Chatelain E, Cannich A, Maitre M, Leste-Lasserre T, Cardinal P, Mendizabal-Zubiaga J, Canduela MJ, Reguero L, Hermans E, Grandes P, Cota D, Marsicano G. Astroglial CB1 cannabinoid receptors regulate leptin signaling in mouse brain astrocytes. Mol Metab 2013; 2:393-404. [PMID: 24327955 DOI: 10.1016/j.molmet.2013.08.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Revised: 07/29/2013] [Accepted: 08/02/2013] [Indexed: 01/22/2023] Open
Abstract
Type-1 cannabinoid (CB1) and leptin (ObR) receptors regulate metabolic and astroglial functions, but the potential links between the two systems in astrocytes were not investigated so far. Genetic and pharmacological manipulations of CB1 receptor expression and activity in cultured cortical and hypothalamic astrocytes demonstrated that cannabinoid signaling controls the levels of ObR expression. Lack of CB1 receptors also markedly impaired leptin-mediated activation of signal transducers and activators of transcription 3 and 5 (STAT3 and STAT5) in astrocytes. In particular, CB1 deletion determined a basal overactivation of STAT5, thereby leading to the downregulation of ObR expression, and leptin failed to regulate STAT5-dependent glycogen storage in the absence of CB1 receptors. These results show that CB1 receptors directly interfere with leptin signaling and its ability to regulate glycogen storage, thereby representing a novel mechanism linking endocannabinoid and leptin signaling in the regulation of brain energy storage and neuronal functions.
Collapse
Key Words
- Astroglial CB1 receptors
- Astroglial leptin receptor
- CB1, type-1 cannabinoid receptor
- Cannabinoid
- Cx, cerebral cortex
- FAAH, fatty acid amide hydrolase
- GFAP, glial fibrillary acidic protein
- Glycogen
- Leptin signaling
- MGL, monoacylglycerol lipase
- ObR, leptin receptor
- ObRb, long-isoform leptin receptor
- P-STAT3, Tyr705-phosphorylated form of STAT3
- P-STAT5, Tyr694-phosphorylated form of STAT5
- STAT3 and 5
- STAT3, transducers and activators of transcription 3
- STAT5, transducers and activators of transcription 5
- VMH, ventromedial hypothalamus
Collapse
Affiliation(s)
- Barbara Bosier
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, F-33000 Bordeaux, France ; University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, F-33000 Bordeaux, France ; Neuropharmacology, Institute of Neurosciences, Université Catholique de Louvain, Av. Hippocrate 54, B1.54.10-10, 1200 Bruxelles, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Enhancement of leptin receptor signaling by SOCS3 deficiency induces development of gastric tumors in mice. Oncogene 2012. [DOI: 10.1038/onc.2012.540] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
34
|
Russo F, Gatta C, De Girolamo P, Cozzi B, Giurisato M, Lucini C, Varricchio E. Expression and immunohistochemical detection of leptin-like peptide in the gastrointestinal tract of the South American sea lion (Otaria flavescens) and the bottlenose dolphin (Tursiops truncatus). Anat Rec (Hoboken) 2012; 295:1482-93. [PMID: 22791650 DOI: 10.1002/ar.22532] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 06/18/2012] [Indexed: 12/31/2022]
Abstract
This study provides an immunohistochemical approach to the expression of leptin in the gastrointestinal tract of the monogastric South American sea lion (Otaria flavescens), and the poligastric bottlenose dolphin (Tursiops truncatus). The specific organization of the gastrointestinal tract is examined in relation to the neuroendocrine regulation of the gut exerted by leptin. In the South American sea lion some leptin-like-immunoreactive (ir) cells, and endocrine type cells, were found in the pit of gastric mucosal folds and in the epithelium of duodenum as well as numerous neurons were detected in the submucosal and myenteric plexuses of the stomach. In the bottlenose dolphin, many leptin-like-ir cells, and exocrine type cells, were identified in the mucosal layer of the main stomach as well as several neurons and nervous fibers were detected in nervous plexuses of main stomach, pyloric stomach, proximal, and middle intestine. Our data suggest that the distribution of leptin-like peptides is similar in the two species, notwithstanding the different anatomical organization of the gastrointestinal apparatus of South American sea lion and bottlenose dolphin. These findings "suggest" the presence of a basal plan in the regulation of food intake, body weight, energy balance and of the gastrointestinal functions in general also in marine mammals with different and specific feeding habits.
Collapse
Affiliation(s)
- Finizia Russo
- Department of Biological, Geological and Environmental Sciences, University of Sannio, Via Port'Arsa 11, 82 100 Benevento, Italy
| | | | | | | | | | | | | |
Collapse
|
35
|
Cesur G, Ozguner F, Yilmaz N, Dundar B. The relationship between ghrelin and adiponectin levels in breast milk and infant serum and growth of infants during early postnatal life. J Physiol Sci 2012; 62:185-90. [PMID: 22311236 PMCID: PMC10717336 DOI: 10.1007/s12576-012-0193-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 01/04/2012] [Indexed: 01/30/2023]
Abstract
Ghrelin and adiponectin have been found in breast milk and are considered to take part in the regulation of growth and energy metabolism of infants. Our aims were to determine ghrelin and adiponectin levels in breast milk and serum samples of mothers and their infants, and to investigate the relationship between their levels and anthropometry of newborn infants during early postnatal life. Total and active ghrelin and adiponectin levels were studied in breast milk, and the serum samples of 25 healthy lactating women and their healthy fullterm infants were taken at the 1st and 4th months of life. Anthropometric measurements of infants were also performed during the study period. Breast milk and infant serum active ghrelin levels were found to be significantly increased at the 4th month of life compared with 1st month levels (p < 0.05). Maternal serum total ghrelin and infant serum adiponectin levels were found to be significantly reduced at the 4th month of life (p < 0.05). Breast milk active ghrelin levels were higher than the infant and maternal serum active ghrelin at the 1st and 4th months (p < 0.05). There was a negative significant correlation between the level of infant serum active ghrelin levels and BMI of infants at the 1st month. A positive significant correlation was found between the level of 1st month infant serum adiponectin levels and weight gain of infants during the study period. Fourth month infant serum adiponectin were also positively correlated with weight and BMI of infants at the 4th month and the weight gain during study period. There was a positive significant correlation between the level of 4th month breast milk active ghrelin and weight gain of infants during the study period. Ghrelin and adiponectin are involved in postnatal growth of infants. Ghrelin in breast milk also seems to be related to the growth of infants during early postnatal life. The sources of these peptides in breast milk are probably both maternal serum and breast tissue itself.
Collapse
Affiliation(s)
- Gokhan Cesur
- Department of Physiology, Adnan Menderes University, Aydin, Turkey.
| | | | | | | |
Collapse
|
36
|
Cammisotto P, Bendayan M. A review on gastric leptin: the exocrine secretion of a gastric hormone. Anat Cell Biol 2012; 45:1-16. [PMID: 22536547 PMCID: PMC3328736 DOI: 10.5115/acb.2012.45.1.1] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/02/2012] [Indexed: 02/08/2023] Open
Abstract
A major advance in the understanding of the regulation of food intake has been the discovery of the adipokine leptin a hormone secreted by the adipose tissue. After crossing the blood-brain barrier, leptin reaches its main site of action at the level of the hypothalamic cells where it plays fundamental roles in the control of appetite and in the regulation of energy expenditure. At first considered as a hormone specific to the white adipose tissue, it was rapidly found to be expressed by other tissues. Among these, the gastric mucosa has been demonstrated to secrete large amounts of leptin. Secretion of leptin by the gastric chief cells was found to be an exocrine secretion. Leptin is secreted towards the gastric lumen into the gastric juice. We found that while secretion of leptin by the white adipose tissue is constitutive, secretion by the gastric cells is a regulated one responding very rapidly to secretory stimuli such as food intake. Exocrine-secreted leptin survives the hydrolytic conditions of the gastric juice by forming a complex with its soluble receptor. This soluble receptor is synthesized by the gastric cells and the leptin-leptin receptor complex gets formed at the level of the gastric chief cell secretory granules before being released into the gastric lumen. The leptin-leptin receptor upon resisting the hydrolytic conditions of the gastric juice is channelled, to the duodenum. Transmembrane leptin receptors expressed at the luminal membrane of the duodenal enterocytes interact with the luminal leptin. Leptin is actively transcytosed by the duodenal enterocytes. From the apical membrane it is transferred to the Golgi apparatus where it binds again its soluble receptor. The newly formed leptin-leptin receptor complex is then secreted baso-laterally into the intestinal mucosa to reach the blood capillaries and circulation thus reaching the hypothalamus where its action regulates food intake. Exocrine-secreted gastric leptin participates in the short term regulation of food intake independently from that secreted by the adipose tissue. Adipose tissue leptin on the other hand, regulates in the long term energy storage. Both tissues work in tandem to ensure management of food intake and energy expenditure.
Collapse
Affiliation(s)
- Philippe Cammisotto
- Department of Pathology and Cell Biology, University of Montreal, Montreal, Quebec, Canada
| | | |
Collapse
|
37
|
Ettore V, Finizia R, Elena C, Giovanni T, David F, Paolo DG, Marina P. Immunohistochemical and immunological detection of ghrelin and leptin in rainbow trout Oncorhynchus mykiss and murray cod Maccullochella peelii peelii as affected by different dietary fatty acids. Microsc Res Tech 2011; 75:771-80. [DOI: 10.1002/jemt.21124] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 10/20/2011] [Indexed: 11/10/2022]
|
38
|
Pan X, Hussain MM. Gut triglyceride production. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:727-35. [PMID: 21989069 DOI: 10.1016/j.bbalip.2011.09.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 10/17/2022]
Abstract
Our knowledge of how the body absorbs triacylglycerols (TAG) from the diet and how this process is regulated has increased at a rapid rate in recent years. Dietary TAG are hydrolyzed in the intestinal lumen to free fatty acids (FFA) and monoacylglycerols (MAG), which are taken up by enterocytes from their apical side, transported to the endoplasmic reticulum (ER) and resynthesized into TAG. TAG are assembled into chylomicrons (CM) in the ER, transported to the Golgi via pre-chylomicron transport vesicles and secreted towards the basolateral side. In this review, we mainly focus on the roles of key proteins involved in uptake and intracellular transport of fatty acids, their conversion to TAG and packaging into CM. We will also discuss intracellular transport and secretion of CM. Moreover, we will bring to light few factors that regulate gut triglyceride production. Furthermore, we briefly summarize pathways involved in cholesterol absorption. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
39
|
Marwarha G, Prasanthi JRP, Schommer J, Dasari B, Ghribi O. Molecular interplay between leptin, insulin-like growth factor-1, and β-amyloid in organotypic slices from rabbit hippocampus. Mol Neurodegener 2011; 6:41. [PMID: 21651786 PMCID: PMC3121598 DOI: 10.1186/1750-1326-6-41] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Accepted: 06/08/2011] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Evidence shows that the insulin-like growth factor-1 (IGF-1) and leptin reduce β-amyloid (Aβ) production and tau phosphorylation, two major hallmarks of Alzheimer's disease (AD). IGF-1 expression involves the JAK/STAT pathway and the expression of leptin is regulated by the mammalian target of rapamycin complex 1 (mTORC1). We have previously shown that Aβ reduces leptin by inhibiting the mTORC1 pathway and Aβ was also suggested to inhibit the JAK/STAT pathway, potentially attenuating IGF-1 expression. As IGF-1 can activate mTORC1 and leptin can modulate JAK/STAT pathway, we determined the extent to which IGF-1 and leptin can upregulate the expression of one another and protect against Aβ-induced downregulation. RESULTS We demonstrate that incubation of organotypic slices from adult rabbit hippocampus with Aβ42 downregulates IGF-1 expression by inhibiting JAK2/STAT5 pathway. Leptin treatment reverses these Aβ42 effects on IGF-1 and treatment with the STAT5 inhibitor completely abrogated the leptin-induced increase in IGF-1. Furthermore, EMSA and ChIP analyses revealed that leptin increases the STAT5 binding to the IGF-1 promoter. We also show that IGF-1 increases the expression of leptin and reverses the Aβ42-induced attenuation in leptin expression via the activation of mTORC1 signaling as the mTORC1 inhibitor rapamycin completely precluded the IGF-1-induced increase in leptin expression. CONCLUSION Our results demonstrate for the first time that Aβ42 downregulates IGF-1 expression and that leptin and IGF-1 rescue one another from downregulation by Aβ42. Our study provides a valuable insight into the leptin/IGF-1/Aβ interplay that may be relevant to the pathophysiology of AD.
Collapse
Affiliation(s)
- Gurdeep Marwarha
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Jaya RP Prasanthi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Jared Schommer
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Bhanu Dasari
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| | - Othman Ghribi
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, 58202, USA
| |
Collapse
|
40
|
Warnakula S, Hsieh J, Adeli K, Hussain MM, Tso P, Proctor SD. New insights into how the intestine can regulate lipid homeostasis and impact vascular disease: frontiers for new pharmaceutical therapies to lower cardiovascular disease risk. Can J Cardiol 2011; 27:183-91. [PMID: 21459267 DOI: 10.1016/j.cjca.2010.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 06/07/2010] [Indexed: 01/27/2023] Open
Abstract
In recent years, evidence has emerged that the intestine is a significant regulator of systemic cholesterol homeostasis and can contribute to raised plasma cholesterol concentration. In this review we provide a context for the role the intestine may have in cardiovascular disease during conditions of chronic disease (insulin resistance, obesity). In particular, we highlight the physiological role of the intestine in lipid absorption, identify novel elements in enterocyte molecular biology, review the concept that chylomicrons and their remnants contribute to atherogenesis during chronic disease, and address new principles of chylomicron overproduction during conditions of insulin resistance including the associated hormonal control of the intestine during these conditions. Finally, we raise the issue of a growing need for novel lipid-lowering pharmaceutical therapies that target intestinal lipid metabolism.
Collapse
|
41
|
Sikalidis AK, Varamini B. Roles of hormones and signaling molecules in describing the relationship between obesity and colon cancer. Pathol Oncol Res 2011; 17:785-90. [PMID: 21221874 DOI: 10.1007/s12253-010-9352-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/17/2010] [Indexed: 01/08/2023]
Abstract
Colon cancer represents a highly prevalent disease in the Western world. While dietary and lifestyle recommendations remain important factors in disease prevention and treatment, epidemiological data have made it clear that obesity and excess body weight remain significant risk factors for the disease. A number of potential direct and indirect relationships exist between obesity and increased risk of colon cancer. Several mechanisms which appear promising and warrant further investigation are discussed here, specifically the modifying role of insulin and insulin-like growth factors, leptin, adipose-tissue induced changes in estrogens and androgens, and inflammatory molecules. A brief review of these hormones and signaling molecules and their action in colon cancer development is described. A thorough integration and understanding of the mechanisms of action these systems exert on colonic epithelia will be important in designing studies and experiments aimed at elucidating disease etiology for prevention and treatment.
Collapse
Affiliation(s)
- Angelos K Sikalidis
- Division of Nutritional Sciences, Cornell University, 214 Savage Hall, Ithaca, NY 14853, USA.
| | | |
Collapse
|
42
|
Childs GV, Akhter N, Haney A, Syed M, Odle A, Cozart M, Brodrick Z, Gaddy D, Suva LJ, Akel N, Crane C, Benes H, Charlesworth A, Luque R, Chua S, Kineman RD. The somatotrope as a metabolic sensor: deletion of leptin receptors causes obesity. Endocrinology 2011; 152:69-81. [PMID: 21084451 PMCID: PMC3033057 DOI: 10.1210/en.2010-0498] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 10/07/2010] [Indexed: 01/01/2023]
Abstract
Leptin, the product of the Lep gene, reports levels of adiposity to the hypothalamus and other regulatory cells, including pituitary somatotropes, which secrete GH. Leptin deficiency is associated with a decline in somatotrope numbers and function, suggesting that leptin may be important in their maintenance. This hypothesis was tested in a new animal model in which exon 17 of the leptin receptor (Lepr) protein was selectively deleted in somatotropes by Cre-loxP technology. Organ genotyping confirmed the recombination of the floxed LepR allele only in the pituitary. Deletion mutant mice showed a 72% reduction in pituitary cells bearing leptin receptor (LEPR)-b, a 43% reduction in LEPR proteins and a 60% reduction in percentages of immunopositive GH cells, which correlated with reduced serum GH. In mutants, LEPR expression by other pituitary cells was like that of normal animals. Leptin stimulated phosphorylated Signal transducer and activator of transcription 3 expression in somatotropes from normal animals but not from mutants. Pituitary weights, cell numbers, IGF-I, and the timing of puberty were not different from control values. Growth curves were normal during the first 3 months. Deletion mutant mice became approximately 30-46% heavier than controls with age, which was attributed to an increase in fat mass. Serum leptin levels were either normal in younger animals or reflected the level of obesity in older animals. The specific ablation of the Lepr exon 17 gene in somatotropes resulted in GH deficiency with a consequential reduction in lipolytic activity normally maintained by GH and increased adiposity.
Collapse
Affiliation(s)
- Gwen V Childs
- Professor and Chair, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Handy JA, Saxena NK, Fu P, Lin S, Mells JE, Gupta NA, Anania FA. Adiponectin activation of AMPK disrupts leptin-mediated hepatic fibrosis via suppressors of cytokine signaling (SOCS-3). J Cell Biochem 2010; 110:1195-207. [PMID: 20564215 PMCID: PMC2907429 DOI: 10.1002/jcb.22634] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adiponectin is an adipocytokine that was recently shown to be anti-fibrogenic in hepatic fibrosis. Leptin, on the other hand, promotes hepatic fibrosis. The purpose of the present study was to elucidate a mechanism (or mechanisms) whereby adiponectin dampens leptin signaling in activated hepatic stellate cells (HSCs), and prevents excess extracellular matrix production. Activated HSCs, between passages 2 and 5, were cultured and exposed to recombinant human adiponectin and recombinant leptin. Immunoblot analysis for SOCS-3, TIMP-1, and the phosphorylated species of Stat3 and adenosine monophosphate-activated protein kinase (AMPK) were conducted. We also examined MMP-1 activity by immunosorbant fluorimetric analysis. In HSCs, adiponectin-induced phosphorylation of AMPK, and subsequently suppressed leptin-mediated Stat3 phosphorylation and SOCS-3 induction. Adiponectin also blocked leptin-stimulated secretion of TIMP-1, and significantly increased MMP-1 activity, in vitro. To extend this study, we treated adiponectin knockout mice (Ad-/-) daily with 5 mg/kg recombinant leptin and/or carbon tetrachloride (2 ml/kg) for 6 weeks. Post-necropsy analysis was performed to examine for inflammation, and histological changes in the Ad-/- and wild-type mice. There was no significant difference in inflammation, or aminotransferases, between mice receiving carbon tetrachloride and leptin versus carbon tetrachloride alone. As anticipated, the combination of leptin and CCl(4) enhanced hepatic fibrosis in both wild-type and Ad-/- mice, as estimated by amount of collagen in injured livers, but wild-type mice had significantly higher levels of SOCS-3 and significantly lower levels of TIMP-1 mRNA and protein than did adiponectin KO mice exposed to both CCl(4) and leptin. We therefore conclude that the protective effects of adiponectin against liver fibrosis require AMPK activation, and may occur through inhibition of the Jak-Stat signal transduction pathway.
Collapse
Affiliation(s)
- Jeffrey A. Handy
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
- FIRST Scholar, Graduate Division of Biomedical and Biological Sciences, Emory University
| | - Neeraj K. Saxena
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
| | - Pingping Fu
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
| | - Songbai Lin
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
| | - Jamie E. Mells
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
- Department of Nutrition and Health Sciences, The Rollins School of Public Health, Emory University
| | - Nitika A. Gupta
- School of Medicine, Department of Pediatrics, Division of Gastroenterology and Nutrition, Emory University
| | - Frank A. Anania
- School of Medicine, Department of Medicine, Division of Digestive Diseases, Emory University
- Department of Nutrition and Health Sciences, The Rollins School of Public Health, Emory University
| |
Collapse
|
44
|
Cammisotto PG, Levy E, Bukowiecki LJ, Bendayan M. Cross-talk between adipose and gastric leptins for the control of food intake and energy metabolism. ACTA ACUST UNITED AC 2010; 45:143-200. [PMID: 20621336 DOI: 10.1016/j.proghi.2010.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2010] [Indexed: 12/25/2022]
Abstract
The understanding of the regulation of food intake has become increasingly complex. More than 20 hormones, both orexigenic and anorexigenic, have been identified. After crossing the blood-brain barrier, they reach their main site of action located in several hypothalamic areas and interact to balance satiety and hunger. One of the most significant advances in this matter has been the discovery of leptin. This hormone plays fundamental roles in the control of appetite and in regulating energy expenditure. In accordance with the lipostatic theory stated by Kennedy in 1953, leptin was originally discovered in white adipose tissue. Its expression by other tissues was later established. Among them, the gastric mucosa has been shown to secrete large amounts of leptin. Both the adipose and the gastric tissues share similar characteristics in the synthesis and storage of leptin in granules, in the formation of a complex with the soluble receptor and a secretion modulated by hormones and energy substrates. However while adipose tissue secretes leptin in a slow constitutive endocrine way, the gastric mucosa releases leptin in a rapid regulated exocrine fashion into the gastric juice. Exocrine-secreted leptin survives the extreme hydrolytic conditions of the gastric juice and reach the duodenal lumen in an intact active form. Scrutiny into transport mechanisms revealed that a significant amount of the exocrine leptin crosses the intestinal wall by active transcytosis. Leptin receptors, expressed on the luminal and basal membrane of intestinal epithelial cells, are involved in the control of nutrient absorption by enterocytes, mucus secretion by goblet cells and motility, among other processes, and this control is indeed different depending upon luminal or basal stimulus. Gastric leptin after transcytosis reaches the central nervous system, to control food intake. Studies using the Caco-2, the human intestinal cell line, in vitro allowed analysis of the mechanisms of leptin actions on the intestinal mucosa, identification of the mechanisms of leptin transcytosis and understanding the modulation of leptin receptors by nutrients and hormones. Exocrine-secreted gastric leptin thus participates in a physiological axis independent in terms of time and regulation from that of adipose tissue to rapidly control food intake and nutrient absorption. Adipocytes and gastric epithelial cells are two cell types the metabolism of which is closely linked to food intake and energy storage. The coordinated secretion of adipose and gastric leptins ensures proper management of food processing and energy storage.
Collapse
Affiliation(s)
- Philippe G Cammisotto
- Department of Pathology and Cell Biology, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, QC, Canada.
| | | | | | | |
Collapse
|
45
|
Hoda RM, Scharl M, Keely SJ, McCole DF, Barrett KE. Apical leptin induces chloride secretion by intestinal epithelial cells and in a rat model of acute chemotherapy-induced colitis. Am J Physiol Gastrointest Liver Physiol 2010; 298:G714-21. [PMID: 20203064 PMCID: PMC2867420 DOI: 10.1152/ajpgi.00320.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The purpose of this study was to investigate whether luminal leptin alters ion transport properties of the intestinal epithelium under acute inflammatory conditions. Monolayers of human intestinal T(84) epithelial cells and a rat model of chemotherapy-induced enterocolitis were used. Cells were treated with leptin and mounted in Ussing chambers to measure basal and secretagogue-induced changes in transepithelial short-circuit current (I(sc)). Furthermore, the role of MAPK and phosphatidylinositol 3-kinase (PI3K) signaling pathways in mediating responses to leptin was investigated. Acute colitis in Sprague-Dawley rats was induced by intraperitoneal injection of 40 mg/kg methotrexate. Leptin (100 ng/ml) induced a time-dependent increase in basal I(sc) in T(84) intestinal epithelial cells (P < 0.01). Moreover, pretreatment of T(84) cells with leptin for up to 1 h significantly potentiated carbachol- and forskolin-induced increases in I(sc). Pretreatment with an inhibitor of MAPK abolished the effect of leptin on basal, carbachol- and forskolin-induced chloride secretion (P < 0.05). However, the PI3K inhibitor, wortmannin, only blunted the effect of leptin on forskolin-induced increases in I(sc). Furthermore, leptin treatment evoked both ERK1/2 and Akt1 phosphorylation in T(84) cells. In the rat model, luminal leptin induced significant increases in I(sc) across segments of proximal and, to a lesser extent, distal colon (P < 0.05). We conclude that luminal leptin is likely an intestinal chloride secretagogue, particularly when present at elevated concentrations and/or in the setting of inflammation. Our findings may provide a mechanistic explanation, at least in part, for the clinical condition of secretory diarrhea both in hyperleptinemic obese patients and in patients with chemotherapy-induced intestinal inflammation.
Collapse
Affiliation(s)
- Raschid M. Hoda
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| | - Michael Scharl
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| | - Stephen J. Keely
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| | - Declan F. McCole
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| | - Kim E. Barrett
- Department of Medicine, University of California, San Diego, School of Medicine, La Jolla, California
| |
Collapse
|
46
|
Receptor-Mediated Transcytosis of Leptin through Human Intestinal Cells In Vitro. Int J Cell Biol 2010; 2010:928169. [PMID: 20454702 PMCID: PMC2862316 DOI: 10.1155/2010/928169] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/11/2010] [Indexed: 01/08/2023] Open
Abstract
Gastric Leptin is absorbed by duodenal enterocytes and released on the basolateral side towards the bloodstream. We investigated in vitro some of the mechanisms of this transport. Caco-2/15 cells internalize leptin from the apical medium and release it through transcytosis in the basal medium in a time- temperature-dependent and saturable fashion. Leptin receptors are revealed on the apical brush-border membrane of the Caco-2 cells. RNA-mediated silencing of the receptor led to decreases in the uptake and basolateral release. Leptin in the basal medium was found bound to the soluble form of its receptor. An inhibitor of clathrin-dependent endocytosis (chlorpromazine) decreased leptin uptake. Confocal immunocytochemistry and the use of brefeldin A and okadaic acid revealed the passage of leptin through the Golgi apparatus. We propose that leptin transcytosis by intestinal cells depends on its receptor, on clathrin-coated vesicles and transits through the Golgi apparatus.
Collapse
|
47
|
Iqbal J, Li X, Chang BHJ, Chan L, Schwartz GJ, Chua SC, Hussain MM. An intrinsic gut leptin-melanocortin pathway modulates intestinal microsomal triglyceride transfer protein and lipid absorption. J Lipid Res 2010; 51:1929-42. [PMID: 20164094 DOI: 10.1194/jlr.m005744] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Fat is delivered to tissues by apoB-containing lipoproteins synthesized in the liver and intestine with the help of an intracellular chaperone, microsomal triglyceride transfer protein (MTP). Leptin, a hormone secreted by adipose tissue, acts in the brain and on peripheral tissues to regulate fat storage and metabolism. Our aim was to identify the role of leptin signaling in MTP regulation and lipid absorption using several mouse models deficient in leptin receptor (LEPR) signaling and downstream effectors. Mice with spontaneous LEPR B mutations or targeted ablation of LEPR B in proopiomelanocortin (POMC) or agouti gene related peptide (AGRP) expressing cells had increased triglyceride in plasma, liver, and intestine. Furthermore, melanocortin 4 receptor (MC4R) knockout mice expressed a similar triglyceride phenotype, suggesting that leptin might regulate intestinal MTP expression through the melanocortin pathway. Mechanistic studies revealed that the accumulation of triglyceride in the intestine might be secondary to decreased expression of MTP and lipid absorption in these mice. Surgical and chemical blockade of vagal efferent outflow to the intestine in wild-type mice failed to alter the triglyceride phenotype, demonstrating that central neural control mechanisms were likely not involved in the observed regulation of intestinal MTP. Instead, we found that enterocytes express LEPR, POMC, AGRP, and MC4R. We propose that a peripheral, local gut signaling mechanism involving LEPR B and MC4R regulates intestinal MTP and controls intestinal lipid absorption.
Collapse
Affiliation(s)
- Jahangir Iqbal
- Department of Cell Biology and Pediatrics, State University of New York Health Science Center at Brooklyn (SUNY Downstate Medical Center), Brooklyn, NY, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Lin Q, Poon SL, Chen J, Cheng L, HoYuen B, Leung PCK. Leptin interferes with 3',5'-cyclic adenosine monophosphate (cAMP) signaling to inhibit steroidogenesis in human granulosa cells. Reprod Biol Endocrinol 2009; 7:115. [PMID: 19849856 PMCID: PMC2770482 DOI: 10.1186/1477-7827-7-115] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Accepted: 10/22/2009] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Obesity has been linked to an increased risk of female infertility. Leptin, an adipocytokine which is elevated during obesity, may influence gonadal function through modulating steroidogenesis in granulosa cells. METHODS The effect of leptin on progesterone production in simian virus 40 immortalized granulosa (SVOG) cells was examined by Enzyme linked immunosorbent assay (ELISA). The effect of leptin on the expression of the steroidogenic enzymes (StAR, P450scc, 3betaHSD) in SVOG cells was examined by real-time PCR and Western blotting. The mRNA expression of leptin receptor isoforms in SVOG cells were examined by using PCR. SVOG cells were co-treated with leptin and specific pharmacological inhibitors to identify the signaling pathways involved in leptin-reduced progesterone production. Silencing RNA against leptin receptor was used to determine that the inhibition of leptin on cAMP-induced steroidogenesis acts in a leptin receptor-dependent manner. RESULTS AND CONCLUSION In the present study, we investigated the cellular mechanisms underlying leptin-regulated steroidogenesis in human granulosa cells. We show that leptin inhibits 8-bromo cAMP-stimulated progesterone production in a concentration-dependent manner. Furthermore, we show that leptin inhibits expression of the cAMP-stimulated steroidogenic acute regulatory (StAR) protein, the rate limiting de novo protein in progesterone synthesis. Leptin induces the activation of ERK1/2, p38 and JNK but only the ERK1/2 (PD98059) and p38 (SB203580) inhibitors attenuate the leptin-induced inhibition of cAMP-stimulated StAR protein expression and progesterone production. These data suggest that the leptin-induced MAPK signal transduction pathway interferes with cAMP/PKA-stimulated steroidogenesis in human granulosa cells. Moreover, siRNA mediated knock-down of the endogenous leptin receptor attenuates the effect of leptin on cAMP-induced StAR protein expression and progesterone production, suggesting that the effect of leptin on steroidogenesis in granulosa cells is receptor dependent. In summary, leptin acts through the MAPK pathway to downregulate cAMP-induced StAR protein expression and progesterone production in immortalized human granulosa cells. These results suggest a possible mechanism by which gonadal steroidogenesis could be suppressed in obese women.
Collapse
Affiliation(s)
- Qing Lin
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, V6H 3V, Canada
- Department of Obstetrics and Gynecology, Beijing Friendship Hospital-Affiliate of Capital University of Medical Sciences, Beijing, PR China
| | - Song Ling Poon
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, V6H 3V, Canada
| | - Junling Chen
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, V6H 3V, Canada
| | - Linan Cheng
- School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Basil HoYuen
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, V6H 3V, Canada
| | - Peter CK Leung
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, V6H 3V, Canada
| |
Collapse
|
49
|
Abstract
AIM To investigate whether change in leptin content of breast milk during lactation acts on neonatal body weight gain. METHODS In total 15 lactating women and their 15 term infants were involved in the study. Breast milk and neonatal serum samples were obtained from the same women and their neonates on the 1st day and any day between the 21st and 30th days after birth. Breast milk and serum leptin concentrations were determined by radioimmunoassay. Anthropometric indexes of the infants were recorded. RESULTS The study was completed with 15 multiparious mothers aged 19-37 years and their infants. The mean collection time of the first samples after birth was 6.07 +/- 1.94 h. The leptin level in the mature milk was significantly higher than in the colostrum (p < 0.001). Neonatal weight and height were significantly increased on 21-30 lactation days compared to 1st day of lactation (p < 0.05 and p < 0.001, respectively). The leptin concentration in the mature milk was negatively correlated with delta BMI (r =-0.53; p < 0.05). The delta breast milk leptin concentration was also found to be inversely correlated with delta BMI (r =-0.529; p < 0.05). CONCLUSION The results of this study have suggested that change in the leptin content of breast milk during lactation might play a role in the regulation of weight gain in healthy neonates.
Collapse
Affiliation(s)
- Hakan Doneray
- Division of Paediatric Endocrinology, Ataturk University, Faculty of Medicine, Erzurum, Turkey.
| | | | | |
Collapse
|
50
|
Hindlet P, Bado A, Kamenicky P, Deloménie C, Bourasset F, Nazaret C, Farinotti R, Buyse M. Reduced intestinal absorption of dipeptides via PepT1 in mice with diet-induced obesity is associated with leptin receptor down-regulation. J Biol Chem 2009; 284:6801-8. [PMID: 19144638 PMCID: PMC2652284 DOI: 10.1074/jbc.m805564200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Revised: 12/26/2008] [Indexed: 12/11/2022] Open
Abstract
Leptin is a major determinant of energy homeostasis, acting both centrally and in the gastrointestinal tract. We previously reported that acute leptin treatment enhances the absorption of di- and tripeptides via the proton-dependent PepT1 transporter. In this study, we investigated the long term effect of leptin on PepT1 levels and activity in Caco2 cell monolayers in vitro. We then assessed the significance of the regulation of PepT1 in vivo in a model of diet-induced obesity. We demonstrated that 1) leptin regulated PepT1 at the transcriptional level, via the MAPK pathway, and at the translational level, via ribosomal protein S6 activation, in Caco2 cells and 2) this activation was systematically followed by a time- and concentration-dependent loss of leptin action reflecting desensitization. Deciphering this desensitization, we demonstrated that leptin induced a down-regulation of its own receptor protein and mRNA expression. More importantly, we showed, in mice with diet-induced obesity, that a 4-week hypercaloric diet resulted in a 46% decrease in PepT1-specific transport, because of a 30% decrease in PepT1 protein and a 50% decrease in PepT1 mRNA levels. As shown in Caco2 cells, these changes in PepT1 were supported by a parallel 2-fold decrease in leptin receptor expression in mice. Taken together, these results indicate that during induction of obesity, leptin resistance may also occur peripherally in the gastrointestinal tract, disrupting the absorption of oligopeptides and peptidomimetic drugs.
Collapse
Affiliation(s)
- Patrick Hindlet
- Laboratoire de Pharmacie Clinique (UPRES EA2706) and IFR-141, Faculté de Pharmacie, Université Paris-Sud 11, 92296 Châtenay-Malabry, France.
| | | | | | | | | | | | | | | |
Collapse
|