This site uses cookies, tags and tracking settings to store information that help give you the very best browsing experience. If you don't change your settings, we'll assume you're happy with this. More information Dismiss this warning

Browse

You are looking at 61 - 70 of 14,371 items for

  • Refine by access: All content x
Clear All
Julia K Panzer Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA

Search for other papers by Julia K Panzer in
Google Scholar
PubMed
Close
and
Alejandro Caicedo Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA

Search for other papers by Alejandro Caicedo in
Google Scholar
PubMed
Close

Long lagging behind insulin, glucagon research has caught up in large part, thanks to technological breakthroughs. Here we review how the field was propelled by the development of novel techniques and approaches. The glucagon radioimmunoassay and islet isolation are methods that now seem trivial, but for decades they were crucial in defining the biology of the pancreatic alpha cell and the role of glucagon in glucose homeostasis. More recently, mouse models have become the main workhorse of this research effort, if not of biomedical research in general. The mouse model allowed detailed mechanistic studies that are revealing alpha cell functions beyond its canonical glucoregulatory role. A recent profusion of gene expression and transcription regulation studies is providing new vistas into what constitutes alpha cell identity. In particular, the combination of transcriptomic techniques with functional recordings promises to move molecular guesswork into real-time physiology. The challenge right now is not to get enamored with these powerful techniques and to make sure that the research continues to be transformative and paradigm shifting. We should imagine a future in which the biology of the alpha cell will be studied at single-cell resolution, non-invasively, and in real time in the human body.

Restricted access
Colin Farquharson Roslin Institute, University of Edinburgh, Midlothian, Edinburgh, UK

Search for other papers by Colin Farquharson in
Google Scholar
PubMed
Close
and
Ruth Andrew University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, UK

Search for other papers by Ruth Andrew in
Google Scholar
PubMed
Close
Restricted access
J Bryce Ortiz Barrow Neurological Institute at Phoenix Children’s Hospital, Phoenix, Arizona, USA
Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA

Search for other papers by J Bryce Ortiz in
Google Scholar
PubMed
Close
,
Sebastian Tellez Arizona State University, School of Life Sciences, Tempe, Arizona, USA

Search for other papers by Sebastian Tellez in
Google Scholar
PubMed
Close
,
Giri Rampal Department of Child Health, University of Arizona College of Medicine, Phoenix, Arizona, USA
Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom

Search for other papers by Giri Rampal in
Google Scholar
PubMed
Close
,
Grant S Mannino Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Grant S Mannino in
Google Scholar
PubMed
Close
,
Nicole Couillard Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Nicole Couillard in
Google Scholar
PubMed
Close
,
Matias Mendez Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Matias Mendez in
Google Scholar
PubMed
Close
,
Tabitha R F Green Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Tabitha R F Green in
Google Scholar
PubMed
Close
,
Sean M Murphy Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Sean M Murphy in
Google Scholar
PubMed
Close
, and
Rachel K Rowe Department of Integrative Physiology, University of Colorado, Boulder, Colorado, USA

Search for other papers by Rachel K Rowe in
Google Scholar
PubMed
Close

Traumatic brain injury (TBI) can damage the hypothalamus and cause improper activation of the growth hormone (GH) axis, leading to growth hormone deficiency (GHD). GHD is one of the most prevalent endocrinopathies following TBI in adults; however, the extent to which GHD affects juveniles remains understudied. We used postnatal day 17 rats (n = 83), which model the late infantile/toddler period, and assessed body weights, GH levels, and number of hypothalamic somatostatin neurons at acute (1, 7 days post injury (DPI)) and chronic (18, 25, 43 DPI) time points. We hypothesized that diffuse TBI would alter circulating GH levels because of damage to the hypothalamus, specifically somatostatin neurons. Data were analyzed with generalized linear and mixed effects models with fixed effects interactions between the injury and time. Despite similar growth rates over time with age, TBI rats weighed less than shams at 18 DPI (postnatal day 35; P = 0.03, standardized effect size [d] = 1.24), which is around the onset of puberty. Compared to shams, GH levels were lower in the TBI group during the acute period (P = 0.196; d = 12.3) but higher in the TBI group during the chronic period (P = 0.10; d = 52.1). Although not statistically significant, TBI-induced differences in GH had large standardized effect sizes, indicating biological significance. The mean number of hypothalamic somatostatin neurons (an inhibitor of GH) positively predicted GH levels in the hypothalamus but did not predict GH levels in the somatosensory cortex. Understanding TBI-induced alterations in the GH axis may identify therapeutic targets to improve the quality of life of pediatric survivors of TBI.

Open access
Jonathan Toledo Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Córdoba, Argentina
Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by Jonathan Toledo in
Google Scholar
PubMed
Close
,
Pablo Aníbal Perez Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Córdoba, Argentina
Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by Pablo Aníbal Perez in
Google Scholar
PubMed
Close
,
Mical Zanetti Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by Mical Zanetti in
Google Scholar
PubMed
Close
,
Graciela Díaz-Torga Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina

Search for other papers by Graciela Díaz-Torga in
Google Scholar
PubMed
Close
,
Jorge Humberto Mukdsi Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Córdoba, Argentina
Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by Jorge Humberto Mukdsi in
Google Scholar
PubMed
Close
, and
Silvina Gutierrez Instituto de Investigaciones en Ciencias de la Salud (INICSA-CONICET), Córdoba, Argentina
Centro de Microscopia Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina

Search for other papers by Silvina Gutierrez in
Google Scholar
PubMed
Close

Due to the current limited knowledge about the role of filamin A (FLNA) in pituitary tumour progression, we aimed to analyse FLNA expression levels and its impact on aggressive markers of pituitary neuroendocrine tumours (PitNETs), using an integrative approach of in vivo and in vitro models and human samples. An increase in the expression levels of FLNA was observed in the advanced tumoural stages of the hyperplastic adenomatous pituitary model, concomitant with a decrease in cell proliferation and with a modification in the subcellular localisation of this protein. Similarly, overexpression of FLNA in the somatolactotropic GH3 cell line induced a decrease in the cell proliferation, promoted a migratory phenotype, increased invasion activity, and decreased the prolactin secretion. Cyclin D1 (CCND1) and cyclin-dependent kinase 4 (CDK4) expression increased in both models in correlation with the increase observed in FLNA levels. When human tissues were analysed a significant increase of FLNA was observed in PitNETs compared to normal pituitary gland, with heterogeneous intracellular localisation. Higher levels of FLNA expression were observed in tumours with invasive characteristics. These results underline the crucial roles of FLNA as a modulator of pathological markers and as a potential prognostic marker in pituitary tumours.

Restricted access
Sebastian R Vanin Departments of Obstetrics and Gynaecology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada

Search for other papers by Sebastian R Vanin in
Google Scholar
PubMed
Close
,
Kendrick Lee Departments of Obstetrics and Gynaecology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada

Search for other papers by Kendrick Lee in
Google Scholar
PubMed
Close
,
Mina Nashed Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada

Search for other papers by Mina Nashed in
Google Scholar
PubMed
Close
,
Brennan Tse Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The Lawson Health Research Institute and Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada

Search for other papers by Brennan Tse in
Google Scholar
PubMed
Close
,
Mohammed Sarikahya Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada

Search for other papers by Mohammed Sarikahya in
Google Scholar
PubMed
Close
,
Sukham Brar Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The Lawson Health Research Institute and Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada

Search for other papers by Sukham Brar in
Google Scholar
PubMed
Close
,
Gregg Tomy Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada

Search for other papers by Gregg Tomy in
Google Scholar
PubMed
Close
,
Amica-Mariae Lucas Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada

Search for other papers by Amica-Mariae Lucas in
Google Scholar
PubMed
Close
,
Thane Tomy Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada

Search for other papers by Thane Tomy in
Google Scholar
PubMed
Close
,
Steven R Laviolette Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada

Search for other papers by Steven R Laviolette in
Google Scholar
PubMed
Close
,
Edith J Arany Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The Lawson Health Research Institute and Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada

Search for other papers by Edith J Arany in
Google Scholar
PubMed
Close
, and
Daniel B Hardy Departments of Obstetrics and Gynaecology, and Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
The Lawson Health Research Institute and the Children's Health Research Institute, London, Ontario, Canada

Search for other papers by Daniel B Hardy in
Google Scholar
PubMed
Close

Reports in North America suggest that up to 20% of young women (18–24 years) use cannabis during pregnancy. This is concerning given clinical studies indicate that maternal cannabis use is associated with fetal growth restriction and dysglycemia in the offspring. Preclinical studies demonstrated that prenatal exposure to Δ9-tetrahydrocannabinol, the main psychoactive component of cannabis, in rat dams led to female-specific deficits in β-cell mass and glucose intolerance/insulin resistance. Yet to date, the contributions of cannabidiol (CBD), the primary nonpsychoactive compound in cannabis, remain elusive. This study aimed to define the effects of in utero cannabidiol (CBD) exposure on postnatal glucose regulation. Pregnant Wistar rat dams received daily intraperitoneal injections of either a vehicle solution or 3 mg/kg of CBD from gestational day (GD) 6 to parturition. CBD exposure did not lead to observable changes in maternal or neonatal outcomes; however, by 3 months of age male CBD-exposed offspring exhibited glucose intolerance despite no changes in pancreatic β/α-cell mass. Transcriptomic analysis on the livers of these CBD-exposed males revealed altered gene expression of circadian rhythm clock machinery, which is linked to systemic glucose intolerance. Furthermore, alterations in hepatic developmental and metabolic processes were also observed, suggesting gestational CBD exposure has a long-lasting detrimental effect on liver health throughout life. Collectively, these results indicate that exposure to CBD alone in pregnancy may be detrimental to the metabolic health of the offspring later in life.

Open access
Medha Sharma Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, India

Search for other papers by Medha Sharma in
Google Scholar
PubMed
Close
,
Yamini Yadav Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, India

Search for other papers by Yamini Yadav in
Google Scholar
PubMed
Close
, and
Chinmoy Sankar Dey Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, New Delhi, India

Search for other papers by Chinmoy Sankar Dey in
Google Scholar
PubMed
Close

Insulin signaling cascade in peripheral insulin-sensitive tissues regulates whole-body glucose metabolism. Any deregulation in this pathway leads to insulin resistance, ultimately leading to metabolic diseases like type 1 diabetes, type 2 diabetes, and obesity. Insulin signaling in the brain has also been studied for many decades and associated with many primary functions like maintenance of synaptic plasticity, regulation of cognition, and circadian rhythm. Importantly, neuronal insulin signaling has also been associated with the regulation of neuronal glucose uptake. Any impairment in neuronal insulin signaling affecting neuronal glucose uptake has been associated with neurodegenerative disorders like Alzheimer’s disease, the process now being termed as type 3 diabetes. Since the criticality lies in proper signaling cascade, determining important points of deregulation is important. In this review, we have discussed some critical points of such deregulation, dividing them into two classes of enzymes: kinases and phosphatases. We have highlighted their individual roles in neuronal insulin signaling, along with their possible implications in neuronal insulin resistance. Future strategies targeting these nodes in neuronal insulin signaling might be helpful in exploring potential therapeutic opportunities to overcome neuronal insulin resistance and related neurodegenerative diseases.

Restricted access
Christy M Gliniak Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States

Search for other papers by Christy M Gliniak in
Google Scholar
PubMed
Close
,
Line Pedersen Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States

Search for other papers by Line Pedersen in
Google Scholar
PubMed
Close
, and
Philipp E Scherer Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, Texas, United States

Search for other papers by Philipp E Scherer in
Google Scholar
PubMed
Close

The prevalence of obesity is increasing exponentially across the globe. The lack of effective treatment options for long-term weight loss has magnified the enormity of this problem. Studies continue to demonstrate that adipose tissue holds a biological memory, one of the most important determinant of long-term weight maintenance. This phenomenon is consistent with the metabolically dynamic role of adipose tissue: it adapts and expands to store for excess energy and serves as an endocrine organ capable of synthesizing a number of biologically active molecules that regulate metabolic homeostasis. An important component of the plasticity of adipose tissue is the extracellular matrix, essential for structural support, mechanical stability, cell signaling and function. Chronic obesity upends a delicate balance of extracellular matrix synthesis and degradation, and the ECM accumulates in such a way that prevents the plasticity and function of the diverse cell types in adipose tissue. A series of maladaptive responses among the cells in adipose tissue leads to inflammation and fibrosis, major mechanisms that explain the link between obesity and insulin resistance, risk of type 2 diabetes, cardiovascular disease, and nonalcoholic fatty liver disease. Adipose tissue fibrosis persists after weight loss and further enhances adipose tissue dysfunction if weight is regained. Here, we highlight the current knowledge of the cellular events governing adipose tissue ECM remodeling during the development of obesity. Our goal is to delineate the relationship more clearly between adipose tissue ECM and metabolic disease, an important step toward better defining the pathophysiology of dysfunctional adipose tissue.

Free access
Russell T Turner Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon, USA

Search for other papers by Russell T Turner in
Google Scholar
PubMed
Close
,
Kenneth A Philbrick Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA

Search for other papers by Kenneth A Philbrick in
Google Scholar
PubMed
Close
,
Carmen P Wong Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA

Search for other papers by Carmen P Wong in
Google Scholar
PubMed
Close
,
Adam J Branscum Biostatistics Program, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA

Search for other papers by Adam J Branscum in
Google Scholar
PubMed
Close
, and
Urszula T Iwaniec Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, USA
Center for Healthy Aging Research, Oregon State University, Corvallis, Oregon, USA

Search for other papers by Urszula T Iwaniec in
Google Scholar
PubMed
Close

Absence of leptin confers metabolic dysfunction resulting in morbid obesity. Bone growth and maturation are also impaired. Partial leptin resistance is more common than leptin deficiency and, when induced by feeding mice a high fat diet, often has a negative effect on bone. Here, we used a genetic model to investigate the skeletal effects of partial and total leptin resistance in mice. This was accomplished by comparing the skeletal phenotypes of 17-week-old female C57Bl6/J wild-type (WT) mice, partial leptin receptor-deficient (db/+) mice and leptin receptor-deficient (db/db) mice (n = 7–8/group), all fed a standard diet. Compared to WT mice, db/db mice were dramatically heavier and hyperleptinemic. These mice were also hypogonadal, hyperglycemic, osteopenic and had lower serum levels of bone turnover markers, osteocalcin and C-terminal telopeptide of type I collagen (CTX). Compared to WT mice, db/+ mice were 14% heavier, had 149% more abdominal white adipose tissue, and were mildly hyperglycemic. db/+ mice did not differ from WT mice in uterine weight or serum levels of markers of bone turnover, although there was a trend for lower osteocalcin. At the bone microarchitectural level, db/+ mice differed from WT mice in having more massive femurs and a trend (P = 0.072) for larger vertebrae. These findings suggest that db/+ mice fed a normal mouse diet compensate for partial leptin resistance by increasing white adipose tissue mass which results in higher leptin levels. Our findings suggest that db/+ mice are a useful diet-independent model for studying the effects of partial leptin resistance on the skeleton.

Restricted access
Seokwon Jo Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA

Search for other papers by Seokwon Jo in
Google Scholar
PubMed
Close
and
Emilyn U Alejandro Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA

Search for other papers by Emilyn U Alejandro in
Google Scholar
PubMed
Close

The metabolic health trajectory of an individual is shaped as early as prepregnancy, during pregnancy, and lactation period. Both maternal nutrition and metabolic health status are critical factors in the programming of offspring toward an increased propensity to developing type 2 diabetes in adulthood. Pancreatic beta-cells, part of the endocrine islets, which are nutrient-sensitive tissues important for glucose metabolism, are primed early in life (the first 1000 days in humans) with limited plasticity later in life. This suggests the high importance of the developmental window of programming in utero and early in life. This review will focus on how changes to the maternal milieu increase offspring’s susceptibility to diabetes through changes in pancreatic beta-cell mass and function and discuss potential mechanisms by which placental-driven nutrient availability, hormones, exosomes, and immune alterations that may impact beta-cell development in utero, thereby affecting susceptibility to type 2 diabetes in adulthood.

Open access
Takanaga Shirai Institute of Health and Sport Sciences, University of Tsukuba, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan
Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo, Japan
Department of Human Sciences, Kanagawa University, Rokkakubashi, Kanagawa-ku, Yokohama-shi, Kanagawa, Japan

Search for other papers by Takanaga Shirai in
Google Scholar
PubMed
Close
,
Tomohiro Iwata Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan

Search for other papers by Tomohiro Iwata in
Google Scholar
PubMed
Close
,
Kazuki Uemichi Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo, Japan
Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan

Search for other papers by Kazuki Uemichi in
Google Scholar
PubMed
Close
,
Riku Tanimura Research Fellow of Japan Society for Promotion Science, Chiyoda-ku, Tokyo, Japan
Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan

Search for other papers by Riku Tanimura in
Google Scholar
PubMed
Close
,
Ryoto Iwai Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan

Search for other papers by Ryoto Iwai in
Google Scholar
PubMed
Close
, and
Tohru Takemasa Institute of Health and Sport Sciences, University of Tsukuba, University of Tsukuba, Tennodai, Tsukuba, Ibaraki, Japan

Search for other papers by Tohru Takemasa in
Google Scholar
PubMed
Close

Graphical abstract

Abstract

Calorie restriction (CR) is a widely recognized dietary approach with beneficial impacts on the entire body, including enhancements in oxidative metabolism and life span extension, while maintaining nutritional balance and calorie intake. However, CR leads to reductions in skeletal muscle and fat mass due to decreased food intake. Consequently, CR significantly modifies the metabolic profile of the entire body and its tissues. The observed benefits in skeletal muscle during CR may be attributed to CR-induced signaling mediators or significant changes in blood profiles associated with CR that regulate homeostasis maintenance. This study aimed to examine the mammalian target of rapamycin signaling and mitochondrial function of skeletal muscle from mice that undergone 8 weeks of CR and cells cultured in their serum to determine whether changes in blood secreted factors during CR affect skeletal muscle cells. C57BL6/J male mice were used. For 8 weeks, these were subjected to ad libitum (AL) or 40% CR. C2C12 myotubes were subsequently treated with media containing 10% mouse serum from AL or CR for 24 h. The results indicated that 8 weeks of CR decreased muscle mass and protein synthesis response compared with the AL group. Interestingly, myotubes conditioned with CR serum exhibited an elevation in the protein synthesis response compared with those treated with AL serum. Furthermore, mitochondrial function was enhanced in both CR mice and cells treated with CR serum. These findings suggest that while CR decreases the protein synthesis response, secretory factors present in the blood during CR can activate protein synthesis and promote mitochondrial function.

Restricted access