Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Tissue Eng Regen Med ; 13(9): 1559-1570, 2019 09.
Article in English | MEDLINE | ID: mdl-31151134

ABSTRACT

Saliva aids in digestion, lubrication, and protection of the oral cavity against dental caries and oropharyngeal infections. Reduced salivary secretion, below an adequate level to sustain normal oral functions, is unfortunately experienced by head and neck cancer patients treated with radiotherapy and by patients with Sjögren's syndrome. No disease-modifying therapies exist to date to address salivary gland hypofunction (xerostomia, dry mouth) because pharmacotherapies are limited by the need for residual secretory acinar cells, which are lost at the time of diagnosis, whereas novel platforms such as cell therapies are yet immature for clinical applications. Autologous salivary gland primary cells have clinical utility as personalized cell therapies, if they could be cultured to a therapeutically useful mass while maintaining their in vivo phenotype. Here, we devised a serum-free scalable suspension culture system that grows partially digested human salivary tissue filtrates composing of acinar and ductal cells attached to their native extracellular matrix components while retaining their 3D in vivo spatial organization; we have coined these salivary spheroids as salivary functional units (SFU). The proposed SFU culture system was sub-optimal, but we have found that the cells could still survive and grow into larger salivary spheroids through cell proliferation and aggregation for 5 to 10 days within the oxygen diffusion rates in vitro. In summary, by using a less disruptive cell isolation procedure as the starting point for primary cell culture of human salivary epithelial cells, we demonstrated that aggregates of cells remained proliferative and continued to express acinar and ductal cell-specific markers.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Epithelial Cells/cytology , Models, Biological , Salivary Glands/cytology , Suspensions , Acinar Cells/cytology , Aquaporin 5/metabolism , Basement Membrane/metabolism , Cell Aggregation , Cell Proliferation , Cell Survival , Culture Media, Serum-Free , Extracellular Matrix/metabolism , Gene Expression Regulation , Humans , Phenotype , Salivary Glands/ultrastructure , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
2.
Anat Rec (Hoboken) ; 301(4): 711-716, 2018 04.
Article in English | MEDLINE | ID: mdl-29236363

ABSTRACT

Recently we reported on the detailed localization of melatonin (and its receptors) in human salivary glands, revealing that serous cells are able to store and secrete melatonin into saliva. Since we found that type 2 diabetic patients display reduced melatonin content in saliva, our next step was to examine the presence of melatonin in salivary glands removed from type 2 diabetic subjects. The resulting data were compared with those previously obtained by identical procedures in non-diabetics, to establish if the diabetic status may affect melatonin distribution. Bioptic samples of diabetic parotid and submandibular glands were fixed, dehydrated, embedded in Epon Resin and processed to demonstrate melatonin reactivity by the immunogold staining method. The labeling density (expressed as the number of gold particles per µm2 /granule) and the percentage of melatonin-positive granules were assessed in diabetic samples. These values were compared with those in non-diabetic samples and differences were evaluated. In parotid and submandibular diabetic glands the reactivity for melatonin was specifically associated with secretory granules and small vesicles in serous cells. Melatonin reactivity was higher in parotid than in submandibular glands. Our data were in line with those obtained in our previous study on non-diabetic glands. Diabetic salivary glands showed a higher labeling density and a lower number of melatonin-positive granules compared to non-diabetic glands. Taken together, these data might explain the decreased salivary melatonin content and the associated oral problems observed in diabetics. Anat Rec, 301:711-716, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Melatonin/metabolism , Salivary Glands/metabolism , Aged , Humans , Immunohistochemistry , Male
3.
J Oral Pathol Med ; 45(7): 510-5, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26694219

ABSTRACT

BACKGROUND: Circulating melatonin is believed to reach body fluids by crossing passively the cell membranes, but alternative ways for melatonin transport also are hypothesized. This investigation was carried out to furnish ultrastructural evidences for melatonin transport by salivary gland cells in order to indicate plausible routes by which circulating melatonin can reach saliva. METHODS: Bioptic samples of parotid, submandibular and labial glands were processed for the electron microscopy and treated to demonstrate melatonin reactivity by the immunogold staining method. RESULTS AND CONCLUSIONS: The preferential sites of melatonin reactivity were the granules and vesicles of serous cells. Our results suggested that the acinar cells are able to store melatonin and that the hormone can be released into saliva through granule and vesicle exocytosis. The quantitative evaluation of labelling showed that the parotid gland is the most involved in the release of melatonin in saliva.


Subject(s)
Melatonin/metabolism , Salivary Glands/metabolism , Acinar Cells/metabolism , Aged , Cell Membrane/metabolism , Exocytosis , Humans , Male , Parotid Gland/metabolism , Saliva/metabolism , Salivary Glands, Minor/metabolism , Submandibular Gland/metabolism
4.
Anat Rec (Hoboken) ; 298(11): 1911-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26264892

ABSTRACT

Type 2 diabetes mellitus represents one of the principal diseases that afflict the world population and is often associated with malfunction of salivary glands and consequent oral diseases. We recently described significant ultrastructural alterations in the human submandibular gland in diabetic patients without evident oral pathologies. Herein, an analogs morphometrical investigation was focused on the parotid gland in order to evaluate if one of the two glands is more affected by diabetes. Parotid fragments from diabetic and nondiabetic patients were fixed, dehydrated, and processed for light and electron microscopy. Serous cells were randomly photographed and the density and size of several structures involved in the secretory process were examined by morphometry. Scanning electron microscopy images revealed significant changes in the number of apically docked granules and vesicles, suggesting that the last steps in exocytosis are somehow altered in diabetic cells. Other variables analyzed by light and transmission electron microscopy such as the size of acini and secretory granules did not show significant changes, but comparison with previous data obtained with submandibular gland cells demonstrated that the two glands are affected differently.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Parotid Gland/pathology , Submandibular Gland/pathology , Humans , Male , Microscopy, Electron, Transmission , Middle Aged , Parotid Gland/ultrastructure , Submandibular Gland/ultrastructure
5.
J Oral Pathol Med ; 44(4): 291-5, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25154984

ABSTRACT

BACKGROUND: Dataon structural alterations in human diabetic salivary glands are scanty and conflicting. The goal of this study is based on the evaluation of the morphological changes in submandibular glands of subjects with well-controlled diabetes and without evident salivary malfunctions. METHODS: Submandibular gland pieces from diabetic and non-diabetic patients were fixed, dehydrated, and processed to obtain sections for light and electron microscopy. Randomly selected micrographs were statistically analyzed to reveal variations in serous acini. RESULTS: Morphometrical evaluation allowed us to reveal significant changes such as enlargement of acinar and granule size, reduction of mitochondrial size, increased density of microbuds and protrusions along luminal membranes. CONCLUSIONS: The results indicate that diabetes affects submandibular gland structure even when glandular function appears unaltered and suggest that morphological changes reflect functional changes chiefly regarding the secretory activity.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Submandibular Gland/pathology , Acinar Cells/pathology , Acinar Cells/ultrastructure , Case-Control Studies , Humans , Lipid Droplets , Male , Microscopy, Electron, Scanning , Middle Aged , Mitochondrial Size , Submandibular Gland/metabolism , Submandibular Gland/ultrastructure
6.
Microsc Res Tech ; 77(10): 790-6, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25044260

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a clinical-pathological syndrome that includes a wide spectrum of morphological alterations. In research, animal models are crucial in evaluating not only the pathogenesis of NAFLD and its progression, but also the therapeutic effects of various agents. Investigations on the ultrastructural features of NAFLD in humans are not copious, due to the difficulty to obtain human samples and to the long time of NAFLD to evolve. Translational comparative studies on the reliability of animal models in representing the histopathologic picture as seen in humans are missing. To overcome this lack of investigations, we compared the ultrastructural NAFLD features of an animal model versus human. Sprague-Dawley rats were fed with a high fat diet (HFD) for 1-4 weeks, while control rats were fed with a standard diet. Human specimens were collected from patients with diagnosed fatty liver disease, undergoing liver biopsies or surgery. Rat and human samples were examined by light microscopy and by transmission and high resolution scanning electron microscopy. The present work demonstrated that NAFLD in animal model and in human, share overlapping ultrastructural features. In conclusion, animal HFD represent an appropriate tool in studying the pathogenesis of NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease/pathology , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Liver/pathology , Liver/ultrastructure , Male , Microscopy , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Middle Aged , Mitochondria, Liver/ultrastructure , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...