Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Exp Biol Med (Maywood) ; 248(12): 1034-1042, 2023 06.
Article in English | MEDLINE | ID: mdl-37073134

ABSTRACT

Recently, several scaffolds have been introduced for urethral tissue engineering. However, acellular human urethral scaffold harvested from deceased donors may provide significant advantages compared to synthetic, composite, or other biological scaffolds. This study aims to develop the protocol for decellularization of the human urethra that preserves substantial extracellular matrix (ECM) components, which are essential for subsequent recellularization mimicking the natural environment of the native ECM. A total of 12 human urethras were harvested from deceased donors. An equal part of every harvested urethra was used as a control sample for analyses. The protocol design was based on the enzyme-detergent-enzyme method. Trypsin and Triton X-100 were used to remove cells, followed by DNase treatment to remove DNA residues. Subsequently, the specimens were continually rinsed in deionized water for seven days. The efficiency of decellularization was determined by histochemistry, immunohistochemical staining, scanning electron microscopy (SEM), and DNA quantification. Histological analysis confirmed cell removal and preservation of urethral structure after decellularization. The preservation of collagen IV and fibronectin was confirmed by histologic examination and immunohistochemical staining. SEM confirmed the maintenance of the ultrastructural architecture of ECM and fibers. DNA content in decellularized urethra was significantly lower compared to the native sample (P < 0.001), and so the criteria for decellularized tissue were met. Cytotoxicity analysis data showed that the matrix-conditioned medium did not contain soluble toxins and had no significant inhibitory effect on cell proliferation, providing evidence that the decellularized samples are not toxic. This study demonstrates the feasibility of the enzyme-detergent-enzyme-based decellularization protocol for removing cellular components and maintaining urethral ECM and its ultrastructure. Moreover, obtained results provide solid ground for recellularization and urethral tissue engineering, which will follow.


Subject(s)
Tissue Engineering , Urethra , Humans , Tissue Engineering/methods , Detergents/pharmacology , Extracellular Matrix/chemistry , DNA , Tissue Scaffolds
2.
Bratisl Lek Listy ; 124(3): 201-204, 2023.
Article in English | MEDLINE | ID: mdl-36598311

ABSTRACT

Human Merkel cells (MCs) were first described by Friedrich S. Merkel in 1875 and named "Tastzellen" (touch cells). Merkel cells are mainly located in the basal layer of the epidermis and are concentrated in touch-sensitive areas. Their density varies among different anatomical sites. Increased concentration was observed in the palms of hands with a predominance in the finger pads and also in the soles and toes. They can be classified according to the function as mechanoreceptive, endocrine, and chemo-sensitive cells. In the development of primary ridges which establish the future fingerprint patterns is assumed that Merkel cells have a significant importance in this process. At about the 7th week EGA, they first time appear in the volar skin and start to occupy the place of future primary ridges at 10 weeks EGA. It will be interesting to study their presence or absence in individuals suffering with abnormal dermatoglyphics and also to study whether the skin diseases associated with altered dermatoglyphics display some deviation regarding the distribution and density of MCs in primary ridges (Fig. 2, Ref. 40). Text in PDF www.elis.sk Keywords: Merkel cells, development, primary ridges, fingerprints, CK-20.


Subject(s)
Merkel Cells , Skin , Humans
3.
Int J Mol Sci ; 23(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36499194

ABSTRACT

This paper presents a proof-of-concept study on the biocolonization of 3D-printed hydroxyapatite scaffolds with mesenchymal stem cells (MSCs). Three-dimensional (3D) printed biomimetic bone structure made of calcium deficient hydroxyapatite (CDHA) intended as a future bone graft was made from newly developed composite material for FDM printing. The biopolymer polyvinyl alcohol serves in this material as a thermoplastic binder for 3D molding of the printed object with a passive function and is completely removed during sintering. The study presents the material, the process of fused deposition modeling (FDM) of CDHA scaffolds, and its post-processing at three temperatures (1200, 1300, and 1400 °C), as well it evaluates the cytotoxicity and biocompatibility of scaffolds with MTT and LDH release assays after 14 days. The study also includes a morphological evaluation of cellular colonization with scanning electron microscopy (SEM) in two different filament orientations (rectilinear and gyroid). The results of the MTT assay showed that the tested material was not toxic, and cells were preserved in both orientations, with most cells present on the material fired at 1300 °C. Results of the LDH release assay showed a slight increase in LDH leakage from all samples. Visual evaluation of SEM confirmed the ideal post-processing temperature of the 3D-printed FDM framework for samples fired at 1300 °C and 1400 °C, with a porosity of 0.3 mm between filaments. In conclusion, the presented fabrication and colonization of CDHA scaffolds have great potential to be used in the tissue engineering of bones.


Subject(s)
Durapatite , Polyvinyl Alcohol , Durapatite/chemistry , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , Porosity
4.
Gen Physiol Biophys ; 41(4): 275-286, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35938961

ABSTRACT

The aim of presented study was to investigate the model of non-invasive method of remote conditioning induced by compression of left forelimb with a tourniquet in three cycles of 2 min of ischemia each followed by 2 min of reperfusion and its influence on the rabbit spinal cord ischemia/reperfusion injury via ubiquitin-mediated stress response. Ubiquitin immunoreaction in spinal cord motor neurons as well as detection of neuronal survival in ventral horns of spinal cord were evaluated. Significantly increased (p < 0.001) number of ubiquitin positive neurons was registered in all remote conditioned groups versus both spinal cord ischemia (SC-ischemia) groups. Our results indicate that remote conditioning significantly attenuated degeneration of motor neurons in all conditioned groups versus SC-ischemia groups in each time point. According to our results, we concluded that the remote conditioning induced by transient limb ischemia is relevant stimulus that provides potent neuroprotection in a model of spinal cord ischemia/ reperfusion injury.


Subject(s)
Reperfusion Injury , Spinal Cord Ischemia , Animals , Ischemia , Rabbits , Reperfusion , Reperfusion Injury/prevention & control , Spinal Cord Ischemia/prevention & control , Ubiquitin
5.
J Mol Histol ; 53(4): 773-780, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35689149

ABSTRACT

The enteric nervous system (ENS), considered as separate branch of the autonomic nervous system, is located throughout the length of the gastrointestinal tract as a series of interconnected ganglionic plexuses. Recently, the ENS is getting more in the focus of gastrointestinal research. For years, the main interest and research was aimed to the enteric neurons and their functional properties in normal conditions, less attention has been paid to the germ-free animals. Germ-free (GF) piglets have clear microbiological background and are reared in sterile environment. GF piglets are regarded as clinically relevant models for studying of human diseases, as these piglets' manifest similar clinical symptoms to humans. In this study we briefly summarised the main characteristics in immunohistochemical distribution of ENS elements in the wall of jejunum and colon of germ-free piglets.


Subject(s)
Enteric Nervous System , Animals , Gastrointestinal Tract , Humans , Neurons , Swine
6.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769175

ABSTRACT

Oral and craniofacial bone defects caused by congenital disease or trauma are widespread. In the case of severe alveolar bone defect, autologous bone grafting has been considered a "gold standard"; however, the procedure has several disadvantages, including limited supply, resorption, donor site morbidity, deformity, infection, and bone graft rejection. In the last few decades, bone tissue engineering combined with stem cell-based therapy may represent a possible alternative to current bone augmentation techniques. The number of studies investigating different cell-based bone tissue engineering methods to reconstruct alveolar bone damage is rapidly rising. As an interdisciplinary field, bone tissue engineering combines the use of osteogenic cells (stem cells/progenitor cells), bioactive molecules, and biocompatible scaffolds, whereas stem cells play a pivotal role. Therefore, our work highlights the osteogenic potential of various dental tissue-derived stem cells and induced pluripotent stem cells (iPSCs), the progress in differentiation techniques of iPSCs into osteoprogenitor cells, and the efforts that have been made to fabricate the most suitable and biocompatible scaffold material with osteoinductive properties for successful bone graft generation. Moreover, we discuss the application of stem cell-derived exosomes as a compelling new form of "stem-cell free" therapy.


Subject(s)
Bone Regeneration , Bone and Bones/metabolism , Induced Pluripotent Stem Cells , Osteogenesis , Tissue Engineering , Tissue Scaffolds/chemistry , Allografts , Animals , Bone Transplantation , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation
7.
Arch Biochem Biophys ; 712: 109031, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34534540

ABSTRACT

Iron, an essential element for most living organism, participates in a wide variety of physiological processes. Disturbance in iron homeostasis has been associated with numerous pathologies, particularly in the heart and brain, which are the most susceptible organs. Under iron-overload conditions, the generation of reactive oxygen species leads to impairment in Ca2+ signaling, fundamentally implicated in cardiac and neuronal physiology. Since iron excess is accompanied by increased expression of iron-storage protein, ferritin, we examined whether ferritin has an effect on the ryanodine receptor - isoform 2 (RYR2), which is one of the major components of Ca2+ signaling. Using the method of planar lipid membranes, we show that ferritin induced an abrupt, permanent blockage of the RYR2 channel. The ferritin effect was strongly voltage dependent and competitively antagonized by cytosolic TEA+, an impermeant RYR2 blocker. Our results collectively indicate that monomeric ferritin highly likely blocks the RYR2 channel by a direct electrostatic interaction within the wider region of the channel permeation pathway.


Subject(s)
Ferritins/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Humans , Lipid Bilayers/metabolism , Male , Membrane Potentials/drug effects , Rats, Wistar , Tetraethylammonium/pharmacology
8.
Biomedicines ; 9(2)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673154

ABSTRACT

Osteoarthritis (OA) belongs to chronic degenerative disorders and is often a leading cause of disability in elderly patients. Typically, OA is manifested by articular cartilage erosion, pain, stiffness, and crepitus. Currently, the treatment options are limited, relying mostly on pharmacological therapy, which is often related to numerous complications. The proper management of the disease is challenging because of the poor regenerative capacity of articular cartilage. During the last decade, cell-based approaches such as implantation of autologous chondrocytes or mesenchymal stem cells (MSCs) have shown promising results. However, the mentioned techniques face their hurdles (cell harvesting, low proliferation capacity). The invention of induced pluripotent stem cells (iPSCs) has created new opportunities to increase the efficacy of the cartilage healing process. iPSCs may represent an unlimited source of chondrocytes derived from a patient's somatic cells, circumventing ethical and immunological issues. Aside from the regenerative potential of iPSCs, stem cell-derived cartilage tissue models could be a useful tool for studying the pathological process of OA. In our recent article, we reviewed the progress in chondrocyte differentiation techniques, disease modeling, and the current status of iPSC-based regenerative therapy of OA.

9.
Int J Mol Sci ; 21(6)2020 Mar 24.
Article in English | MEDLINE | ID: mdl-32213859

ABSTRACT

Huntington's disease (HD) is an inherited, autosomal dominant, degenerative disease characterized by involuntary movements, cognitive decline, and behavioral impairment ending in death. HD is caused by an expansion in the number of CAG repeats in the huntingtin gene on chromosome 4. To date, no effective therapy for preventing the onset or progression of the disease has been found, and many symptoms do not respond to pharmacologic treatment. However, recent results of pre-clinical trials suggest a beneficial effect of stem-cell-based therapy. Induced pluripotent stem cells (iPSCs) represent an unlimited cell source and are the most suitable among the various types of autologous stem cells due to their patient specificity and ability to differentiate into a variety of cell types both in vitro and in vivo. Furthermore, the cultivation of iPSC-derived neural cells offers the possibility of studying the etiopathology of neurodegenerative diseases, such as HD. Moreover, differentiated neural cells can organize into three-dimensional (3D) organoids, mimicking the complex architecture of the brain. In this article, we present a comprehensive review of recent HD models, the methods for differentiating HD-iPSCs into the desired neural cell types, and the progress in gene editing techniques leading toward stem-cell-based therapy.


Subject(s)
Huntington Disease/genetics , Induced Pluripotent Stem Cells/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Differentiation , Genetic Therapy/methods , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Huntington Disease/therapy , Induced Pluripotent Stem Cells/cytology , Organoids/metabolism , Organoids/pathology
10.
Exp Clin Endocrinol Diabetes ; 128(5): 339-346, 2020 May.
Article in English | MEDLINE | ID: mdl-30114722

ABSTRACT

Diabetes type 1 (T1D) is a common autoimmune disease characterized by permanent destruction of the insulin-secreting ß-cells in pancreatic islets, resulting in a deficiency of the glucose-lowering hormone insulin and persisting high blood glucose levels. Insulin has to be replaced by regular subcutaneous injections, and blood glucose level must be monitored due to the risk of hyperglycemia. Recently, transplantation of new pancreatic ß-cells into T1D patients has come to be considered one of the most potentially effective treatments for this disease. Therefore, much effort has focused on understanding the regulation of ß-cells. Induced pluripotent stem cells (iPSCs) represent a valuable source for T1D modelling and cell replacement therapy because of their ability to differentiate into all cell types in vitro. Recent advances in stem cell-based therapy and gene-editing tools have enabled the generation of functionally adult pancreatic ß-cells derived from iPSCs. Although animal and human pancreatic development and ß-cell physiology have significant differences, animal models represent an important tool in evaluating the therapeutic potential of iPSC-derived ß-cells on type 1 diabetes treatment. This review outlines the recent progress in iPSC-derived ß-cell differentiation methods, disease modelling, and future perspectives.


Subject(s)
Cell- and Tissue-Based Therapy , Diabetes Mellitus, Type 1/therapy , Induced Pluripotent Stem Cells , Insulin-Secreting Cells/transplantation , Animals , Humans
11.
Int J Mol Sci ; 20(15)2019 Aug 06.
Article in English | MEDLINE | ID: mdl-31390782

ABSTRACT

Regeneration of injuries occurring in the central nervous system, particularly spinal cord injuries (SCIs), is extremely difficult. The complex pathological events following a SCI often restrict regeneration of nervous tissue at the injury site and frequently lead to irreversible loss of motor and sensory function. Neural stem/progenitor cells (NSCs/NPCs) possess neuroregenerative and neuroprotective features, and transplantation of such cells into the site of damaged tissue is a promising stem cell-based therapy for SCI. However, NSC/NPCs have mostly been induced from embryonic stem cells or fetal tissue, leading to ethical concerns. The pioneering work of Yamanaka and colleagues gave rise to the technology to induce pluripotent stem cells (iPSCs) from somatic cells, overcoming these ethical issues. The advent of iPSCs technology has meant significant progress in the therapy of neurodegenerative disease and nerve tissue damage. A number of published studies have described the successful differentiation of NSCs/NPCs from iPSCs and their subsequent engraftment into SCI animal models, followed by functional recovery of injury. The aim of this present review is to summarize various iPSC- NPCs differentiation methods, SCI modelling, and the current status of possible iPSC- NPCs- based therapy of SCI.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Nerve Regeneration , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/therapy , Stem Cell Transplantation , Animals , Humans , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Spinal Cord Injuries/etiology , Tissue Scaffolds
12.
Medicina (Kaunas) ; 55(2)2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30781716

ABSTRACT

In recent years, the interstitial cells telocytes, formerly known as interstitial Cajal-like cells, have been described in almost all organs of the human body. Although telocytes were previously thought to be localized predominantly in the organs of the digestive system, as of 2018 they have also been described in the lymphoid tissue, skin, respiratory system, urinary system, meninges and the organs of the male and female genital tracts. Since the time of eminent German pathologist Rudolf Virchow, we have known that many pathological processes originate directly from cellular changes. Even though telocytes are not widely accepted by all scientists as an individual and morphologically and functionally distinct cell population, several articles regarding telocytes have already been published in such prestigious journals as Nature and Annals of the New York Academy of Sciences. The telocyte diversity extends beyond their morphology and functions, as they have a potential role in the etiopathogenesis of different diseases. The most commonly described telocyte-associated diseases (which may be best termed "telocytopathies" in the future) are summarized in this critical review. It is difficult to imagine that a single cell population could be involved in the pathogenesis of such a wide spectrum of pathological conditions as extragastrointestinal stromal tumors ("telocytomas"), liver fibrosis, preeclampsia during pregnancy, tubal infertility, heart failure and psoriasis. In any case, future functional studies of telocytes in vivo will help to understand the mechanism by which telocytes contribute to tissue homeostasis in health and disease.


Subject(s)
Homeostasis/physiology , Interstitial Cells of Cajal/pathology , Telocytes/pathology , Antigens, CD34/immunology , Humans , Immunophenotyping , Interstitial Cells of Cajal/immunology , Neovascularization, Physiologic , Receptor, Platelet-Derived Growth Factor alpha/immunology , Receptor, Platelet-Derived Growth Factor beta/immunology , Regeneration , Signal Transduction , Telocytes/immunology
13.
Eur J Histochem ; 62(1): 2836, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29569875

ABSTRACT

Human Merkel cells (MCs) were first described by Friedrich S. Merkel in 1875 and named "Tastzellen" (touch cells). Merkel cells are primarily localized in the basal layer of the epidermis and concentrated in touch-sensitive areas. In our previous work, we reported on the distribution of MCs in the human esophagus, so therefore we chose other parts of the human body to study them. We selected the human vagina, because it has a similar epithelium as the esophagus and plays very important roles in reproduction and sexual pleasure. Due to the fact that there are very few research studies focusing on the innervation of this region, we decided to investigate the occurrence of MCs in the anterior wall of the vagina. The aim of our research was to identify MCs in the stratified squamous non-keratinized epithelium of the human vagina in 20 patients. For the identification of Merkel cells by light microscopy, we used antibodies against simple-epithelial cytokeratins (especially anti-cytokeratin 20). We also tried to identify them using transmission electron microscopy. Our investigation confirmed that 10 (50 %) of 20 patients had increased number of predominantly intraepithelial CK20 positive "Merkel-like" cells (MLCs) in the human vaginal epithelium. Subepithelial CK20 positive MLCs were observed in only one patient (5%). We tried to identify them also using transmission electron microscopy. Our investigation detected some unique cells that may be MCs. The purpose of vaginal innervation is still unclear. There are no data available concerning the distribution of MCs in the human vagina, so it would be interesting to study the role of MCs in the vaginal epithelium, in the context of innervation and epithelial biology.


Subject(s)
Epithelium/chemistry , Merkel Cells/chemistry , Vagina/chemistry , Adult , Aged , Aged, 80 and over , Epithelium/ultrastructure , Female , Humans , Immunohistochemistry , Merkel Cells/ultrastructure , Microscopy, Electron, Transmission , Middle Aged , Vagina/ultrastructure
14.
Med Hypotheses ; 110: 105-109, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29317051

ABSTRACT

Heart failure is the end stage of cardiovascular abnormalities. Studies have primarily focused on the functional changes of cardiomyocytes in the failing heart from different animal models with very little information in the human condition. In addition little is known about the ultrastructural changes that proceed in cardiomyocytes in route to failure. The aim of this study was to examine the ultrastructural changes in the myocardium of human with end-stage heart failure. Left ventricular myocardial tissue samples from 7 patients with end-stage heart failure were examined with transmission and scanning electron microscopy. All heart failure patients were of New York Heart Association (NYHA) class III-IV. The data indicated normal three-dimensional arrangement of cardiac muscle cells in failing myocardium. The various organelles in cardiomyocytes including the nucleus, mitochondria, myofibrils, T-tubules and intercalated discs did not exhibit any remarkable morphological changes. We did observe the appearance of small membrane bound vesicles which appear to be associated with the intercalated discs. The nearly normal ultrastructure and arrangement of cardiomyocytes was remarkable in contrast to the dramatic clinical status of these patients in heart failure. These observations support the hypothesis, that there are no dramatic changes in the ultrastructure or three-dimensional architecture of cardiomyocytes in end-stage failing human myocardium.


Subject(s)
Heart Failure/pathology , Myocytes, Cardiac/ultrastructure , Adult , Animals , Female , Heart Failure/physiopathology , Heart Ventricles/ultrastructure , Humans , Intercellular Junctions/ultrastructure , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Middle Aged , Mitochondria, Heart/ultrastructure , Models, Cardiovascular , Myocardium/ultrastructure , Myofibrils/ultrastructure
15.
Med Hypotheses ; 110: 64-67, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29317071

ABSTRACT

Uterine telocytes are interstitial cells characterized by a very long cytoplasmic prolongations, which form a 3D network, functionally integrating a wide variety of different cells. Leiomyomas (uterine fibroids) are benign tumors, which pose a huge threat concerning various health problems in women affected by this condition. The exact cause of leiomyomas development is, however, still largely unknown. Therefore, in an attempt to clarify their etiology, we performed an immunohistochemical characterization of telocytes in leiomyomas as well as in normal myometrium. Tissue samples of intramural leiomyomas from 26 women (age 46.26 ±â€¯11.07) were immunohistochemically stained for the expression of c-kit (CD117) antigen, one of the markers of telocytes. C-kit (CD117) antigen is useful for a routine immunohistochemical identification of uterine telocytes in histological sections of myometrium. In normal, healthy myometrium the c-kit positive telocytes occupy approximately 2.2% of the area of a tissue slide, contrasting with no detectable c-kit positive cells within leiomyomas. As telocytes are thought to be key players in the regulation of tissue homoeostasis, our data suggest that uterine telocyte loss may have important implications in the pathogenesis of leiomyomas. In addition, we supposed to summarize three hypotheses on the association of the cells telocytes loss within the myometrium and formation of leiomyomas. These hypotheses include the loss of telocytes' functions as "sex hormone sensors" and regulators of smooth muscle cells cycle; the role of telocytes as progenitor cells for the development of leiomyomas; and the hypothesis of decreased angiogenesis after telocytes' loss with subsequent hypoxia (as a key factor for leiomyomas development).


Subject(s)
Leiomyoma/etiology , Leiomyoma/pathology , Telocytes/pathology , Uterine Neoplasms/etiology , Uterine Neoplasms/pathology , Adult , Aged , Female , Humans , Immunohistochemistry , Leiomyoma/metabolism , Middle Aged , Models, Biological , Myometrium/metabolism , Myometrium/pathology , Proto-Oncogene Proteins c-kit/metabolism , Telocytes/metabolism , Uterine Neoplasms/metabolism , Young Adult
16.
Cell Tissue Bank ; 19(1): 61-68, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28812172

ABSTRACT

Acellular dermal matrix (ADM) is a tissue graft of allogeneic origin from post-mortem tissue donors prepared by an innovative decellularization process. The newly developed non-toxic and low cost decellularization process of cadaver origin dermis included ADM in breast reconstruction procedures proved to help coverage of the lower-pole of breast expanders or implants. As the results have shown, it did help to eliminate autologous dermis donor site morbidity along with shortening the operation time by avoiding elevation of additional muscle or fascia during the operation. Main aims of this article include histology evaluation of allogeneic acellular dermal matrix prepared by a new decellularization method and presentation of clinical results of its use. A total of 22 patients underwent 26 ADM based breast reconstructions. The mean patient's follow up was 12.6 months. Average total size of ADM used for one breast was 273 cm2. Post-operative complications occurred in 3 patients including one expander infection, one expander extrusion and one expander pocket disfiguration. Microscopic analysis of tissue samples has confirmed incorporation of the acellular dermal matrices into the surrounding connective tissue without any noticeable immune reaction. In a majority of the ADM samples we found pseudocapsullar formation on implant side of samples without acute or chronic inflammatory cells. The use of ADM prepared by new preparation method in expansive post mastectomy breast reconstruction was associated by a relatively low complication rate resulting in good outcomes.


Subject(s)
Acellular Dermis , Mammaplasty/methods , Mastectomy , Adult , Aged , Breast Implants , Humans , Middle Aged , Retrospective Studies , Tissue Expansion Devices , Treatment Outcome
17.
J Adv Res ; 8(4): 321-327, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28386481

ABSTRACT

Bone disorders are a group of varied acute and chronic traumatic, degenerative, malignant or congenital conditions affecting the musculoskeletal system. They are prevalent in society and, with an ageing population, the incidence and impact on the population's health is growing. Severe persisting pain and limited mobility are the major symptoms of the disorder that impair the quality of life in affected patients. Current therapies only partially treat the disorders, offering management of symptoms, or temporary replacement with inert materials. However, during the last few years, the options for the treatment of bone disorders have greatly expanded, thanks to the advent of regenerative medicine. Skeletal cell-based regeneration medicine offers promising reparative therapies for patients. Mesenchymal stem (stromal) cells from different tissues have been gradually translated into clinical practice; however, there are a number of limitations. The introduction of reprogramming methods and the subsequent production of induced pluripotent stem cells provides a possibility to create human-specific models of bone disorders. Furthermore, human-induced pluripotent stem cell-based autologous transplantation is considered to be future breakthrough in the field of regenerative medicine. The main goal of the present paper is to review recent applications of induced pluripotent stem cells in bone disease modeling and to discuss possible future therapy options. The present article contributes to the dissemination of scientific and pre-clinical results between physicians, mainly orthopedist and thus supports the translation to clinical practice.

18.
Ann Anat ; 211: 140-148, 2017 May.
Article in English | MEDLINE | ID: mdl-28279759

ABSTRACT

Hassall's corpuscles are the most prominent structures in the human thymus. However, relatively few analyses have been performed to determine their function and cellular origins during development. In this study, we evaluated the cellular microenvironment of human thymic Hassall's corpuscles using histochemistry, immunohistochemistry, and transmission electron microscopy. We examined 95 human thymic tissue samples, which were perioperatively obtained from children undergoing cardiac surgery. To characterize the complex cellular microenvironment of human thymic corpuscles, we used a panel of 14 different antibodies to identify discrete cell types. We also utilized various histochemical methods (PAS reaction, alcian blue staining, alkaline phosphatase and acid phosphatase activity staining, von Kossa staining of calcified particles) and transmission electron microscopy to visualize these structures. Considerable variation in the sizes, shapes, and numbers of Hassall's corpuscles was observed, even amongst children of the same age. Inside the largest Hassall's corpuscles, cystic dilatation with an accumulation of cellular debris was found. These morphological observations might be associated with disruptions in the formation, migration, or differentiation of cardiac neural crest cells, which are essential for heart and thymus development. Immunohistochemical staining and electron microscopy revealed that Hassall's corpuscles resemble other types of stratified squamous epithelia. Most Hassall's corpuscles are heterocellular, consisting of thymic epithelial cells, macrophages, interdigitating dendritic cells, myoid cells, and, occasionally, mast cells and lymphocytes. To explore the potential functions of Hassall's corpuscles, we found that the concentrations of B-lymphocytes and BCL2-positive lymphocytes suggested a role in regulation of lymphopoiesis. We also found that these structures do not originate from the perivascular epithelium as previously proposed, nor could we identify blood or lymph endothelial cells in close proximity. This leaves the origins of Hassall's corpuscles an open question.


Subject(s)
Epithelial Cells/ultrastructure , Lymphocytes/ultrastructure , Thymus Gland/ultrastructure , Cellular Microenvironment , Child , Child, Preschool , Epithelial Cells/chemistry , Female , Humans , Lymphocytes/chemistry , Male , Reproducibility of Results , Sensitivity and Specificity , Thymus Gland/chemistry
19.
Cell Tissue Bank ; 18(1): 45-52, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27873029

ABSTRACT

A novel approach for stem cell generation is the attempt to induce conversion of the adult somatic cells into pluripotent stem cells so called induced pluripotent stem cells (iPSCs) by introducing specific transcription factors. iPSCs have two essential cell characteristics, they are pluripotent and posses long term cell-renewal capacity. Additionally, iPSCs can be derived from patient-specific somatic cells, thus bypassing ethical and immunological issues. The aim of our study was to reprogram long-term cryopreserved human neonatal fibroblasts by new method using lipid nano-particle technology (Lipofectamine 3000 reagent transfection system) in combination with Epi 5 reprogramming vectors. Obtained iPSCs were characterized by several sophisticated methods of molecular biology and microscopy. Distinct colonies of iPSCs started to appear by day 20 after reprogramming. The presence of iPSCs colonies was proved by alkaline phosphatase (AP) live staining. After manual picking the colonies and their subsequent passaging, they did not lose ability to form embryoid bodies, they were positive for AP, Tra-1-60, and SSEA-5. Moreover, obtained iPSCs expressed pluripotency markers Oct4, Sox2 and Nanog, and the expression levels of chondrogenic, osteogenic and adipogenic markers were significantly higher in comparison to control (p < 0.05). In summary, we have demonstrated that long-term cryopreserved human neonatal fibroblasts can be reprogrammed into iPSCs and after further analysis concerns on their biological safety they may be used as patient-specific cells in regenerative medicine.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Line , Cryopreservation , Fibroblasts/metabolism , Genetic Vectors/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Lipids/chemistry , Plasmids/genetics
20.
Can J Physiol Pharmacol ; 94(11): 1117-1121, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27428566

ABSTRACT

Key morphological discoveries in recent years have included the discovery of new cell populations inside the heart called cardiac telocytes. These newly described cells of the connective tissue have extremely long cytoplasmic processes through which they form functionally connected three-dimensional networks that connect cells of the immune system, nerve fibers, cardiac stem cells, and cardiac muscle cells. Based on their functions, telocytes are also referred to as "connecting cells" or "nurse cells" for cardiac progenitor stem cells. In this critical review, we provide a summary of the latest research on cardiac telocytes localized in all layers of the heart - from the historical background of their discovery, through ultrastructural, immunohistochemical, and functional characterizations, to the application of this knowledge to the fields of cardiology, stem cell research, and regenerative medicine.

SELECTION OF CITATIONS
SEARCH DETAIL
...