In recent times, https://stemcellscosts.com/ stem cell therapy has turn out to be a really promising and advanced scientific research subject. The event of remedy strategies has evoked great expectations. This paper is a evaluate focused on the discovery of different stem cells and the potential therapies based mostly on these cells. The genesis of stem cells is adopted by laboratory steps of managed stem cell culturing and derivation. Quality management and teratoma formation assays are vital procedures in assessing the properties of the stem cells examined. Derivation methods and the utilization of culturing media are essential to set proper environmental conditions for controlled differentiation. Among many kinds of stem tissue functions, using graphene scaffolds and the potential of extracellular vesicle-primarily based therapies require attention because of their versatility. The review is summarized by challenges that stem cell therapy should overcome to be accepted worldwide. A wide variety of potentialities makes this innovative therapy a turning level in modern medicine, providing hope for untreatable diseases.
Stem cell classification
Stem cells are unspecialized cells of the human body. They’re able to differentiate into any cell of an organism and have the power of self-renewal. Stem cells exist each in embryos and grownup cells. There are a number of steps of specialization. Developmental potency is lowered with each step, which means that a unipotent stem cell is not in a position to differentiate into as many types of cells as a pluripotent one. This chapter will concentrate on stem cell classification to make it simpler for the reader to comprehend the following chapters.
Totipotent stem cells are able to divide and differentiate into cells of the entire organism. Totipotency has the very best differentiation potential and permits cells to kind both embryo and additional-embryonic constructions. One instance of a totipotent cell is a zygote, which is formed after a sperm fertilizes an egg. These cells can later develop either into any of the three germ layers or kind a placenta. After approximately 4 days, the blastocyst’s internal cell mass becomes pluripotent. This structure is the supply of pluripotent cells.
Pluripotent stem cells (PSCs) kind cells of all germ layers however not extraembryonic buildings, such as the placenta. Embryonic stem cells (ESCs) are an example. ESCs are derived from the inner cell mass of preimplantation embryos. Another instance is induced pluripotent stem cells (iPSCs) derived from the epiblast layer of implanted embryos. Their pluripotency is a continuum, beginning from completely pluripotent cells similar to ESCs and iPSCs and ending on representatives with much less potency-multi-, oligo- or unipotent cells. One of many strategies to evaluate their exercise and spectrum is the teratoma formation assay. iPSCs are artificially generated from somatic cells, they usually function equally to PSCs. Their culturing and utilization are very promising for current and future regenerative medication.
Multipotent stem cells have a narrower spectrum of differentiation than PSCs, however they will specialize in discrete cells of specific cell lineages. One example is a haematopoietic stem cell, which can turn into several forms of blood cells. After differentiation, a haematopoietic stem cell turns into an oligopotent cell. Its differentiation skills are then restricted to cells of its lineage. However, some multipotent cells are capable of conversion into unrelated cell sorts, which suggests naming them pluripotent cells.
Oligopotent stem cells can differentiate into several cell varieties. A myeloid stem cell is an example that can divide into white blood cells however not crimson blood cells.
Unipotent stem cells are characterized by the narrowest differentiation capabilities and a particular property of dividing repeatedly. Their latter function makes them a promising candidate for therapeutic use in regenerative medicine. These cells are solely able to type one cell type, e.g. dermatocytes.
Stem cell biology
A blastocyst is formed after the fusion of sperm and ovum fertilization. Its interior wall is lined with brief-lived stem cells, specifically, embryonic stem cells. Blastocysts are composed of two distinct cell sorts: the inner cell mass (ICM), which develops into epiblasts and induces the event of a foetus, and the trophectoderm (TE). Blastocysts are liable for the regulation of the ICM microenvironment. The TE continues to develop and forms the extraembryonic assist constructions needed for the profitable origin of the embryo, such as the placenta. As the TE begins to type a specialised assist structure, the ICM cells remain undifferentiated, totally pluripotent and proliferative [1]. The pluripotency of stem cells allows them to type any cell of the organism. Human embryonic stem cells (hESCs) are derived from the ICM. During the strategy of embryogenesis, cells form aggregations called germ layers: endoderm, mesoderm and ectoderm (Fig. 1), each finally giving rise to differentiated cells and tissues of the foetus and, later on, the grownup organism [2]. After hESCs differentiate into one of the germ layers, they develop into multipotent stem cells, whose potency is proscribed to only the cells of the germ layer. This process is short in human improvement. After that, pluripotent stem cells occur all over the organism as undifferentiated cells, and their key abilities are proliferation by the formation of the next era of stem cells and differentiation into specialised cells under certain physiological conditions.
Oocyte development and formation of stem cells: the blastocoel, which is formed from oocytes, consists of embryonic stem cells that later differentiate into mesodermal, ectodermal, or endodermal cells. Blastocoel develops into the gastrula
Signals that influence the stem cell specialization process might be divided into exterior, corresponding to physical contact between cells or chemical secretion by surrounding tissue, and inside, that are indicators controlled by genes in DNA.
Stem cells also act as inner repair systems of the body. The replenishment and formation of new cells are unlimited so long as an organism is alive. Stem cell exercise relies on the organ by which they’re in; for example, in bone marrow, their division is constant, although in organs such as the pancreas, division only happens under particular physiological circumstances.
Stem cell practical division
Whole-physique growth
During division, the presence of different stem cells is dependent upon organism improvement. Somatic stem cell ESCs can be distinguished. Although the derivation of ESCs without separation from the TE is possible, such a mixture has development limits. Because proliferating actions are limited, co-tradition of those is normally averted.
ESCs are derived from the inside cell mass of the blastocyst, which is a stage of pre-implantation embryo ca. Four days after fertilization. After that, these cells are placed in a culture dish crammed with culture medium. Passage is an inefficient however popular technique of sub-culturing cells to other dishes. These cells will be described as pluripotent because they’re able to ultimately differentiate into every cell kind within the organism. Since the beginning of their studies, there have been moral restrictions linked to the medical use of ESCs in therapies. Most embryonic stem cells are developed from eggs which were fertilized in an in vitro clinic, not from eggs fertilized in vivo.
Somatic or grownup stem cells are undifferentiated and found amongst differentiated cells in the whole physique after growth. The function of these cells is to allow the healing, progress, and substitute of cells which are misplaced every day. These cells have a restricted range of differentiation options. Among many sorts, there are the following:
– Mesenchymal stem cells are present in many tissues. In bone marrow, these cells differentiate primarily into the bone, cartilage, and fat cells. As stem cells, they are an exception as a result of they act pluripotently and can specialize in the cells of any germ layer.
– Neural cells give rise to nerve cells and their supporting cells-oligodendrocytes and astrocytes.
– Haematopoietic stem cells type all sorts of blood cells: pink, white, and platelets.
– Skin stem cells kind, for instance, keratinocytes, which type a protecting layer of skin.
The proliferation time of somatic stem cells is longer than that of ESCs. It is possible to reprogram grownup stem cells again to their pluripotent state. This can be carried out by transferring the grownup nucleus into the cytoplasm of an oocyte or by fusion with the pluripotent cell. The identical approach was used throughout cloning of the famous Dolly sheep.
hESCs are involved in whole-body improvement. They can differentiate into pluripotent, totipotent, multipotent, and unipotent cells (Fig. 2) [2].
Changes within the potency of stem cells in human body improvement. Potency ranges from pluripotent cells of the blastocyst to unipotent cells of a particular tissue in a human body such because the skin, CNS, or bone marrow. Reversed pluripotency might be achieved by the formation of induced pluripotent stem cells utilizing both octamer-binding transcription factor (Oct4), intercourse-figuring out region Y (Sox2), Kruppel-like factor 4 (Klf4), or the Myc gene
Pluripotent cells could be named totipotent if they can moreover form extraembryonic tissues of the embryo. Multipotent cells are restricted in differentiating to each cell sort of given tissue. When tissue accommodates only one lineage of cells, stem cells that kind them are called either called oligo- or unipotent.
iPSC quality management and recognition by morphological differences
The comparability of stem cell strains from completely different people is required for iPSC traces to be utilized in therapeutics [3]. Among crucial high quality procedures, the following could be distinguished:
Short tandem repeat analysis-This is the comparison of specific loci on the DNA of the samples. It’s used in measuring an exact number of repeating units. One unit consists of two to 13 nucleotides repeating many occasions on the DNA strand. A polymerase chain response is used to check the lengths of short tandem repeats. The genotyping process of source tissue, cells, and iPSC seed and grasp cell banks is really useful.
Identity evaluation-The unintentional switching of traces, resulting in different stem cell line contamination, requires rigorous assay for cell line identification.
Residual vector testing-An appearance of reprogramming vectors integrated into the host genome is hazardous, and testing their presence is a obligatory process. It is a commonly used process for generating excessive-high quality iPSC traces. An acceptable threshold in excessive-high quality research-grade iPSC line collections is ≤ 1 plasmid copies per a hundred cells. During the procedure, 2 different regions, common to all plasmids, ought to be used as specific targets, akin to EBNA and CAG sequences [3]. To precisely signify the test reactions, a standard curve needs to be prepared in a provider of gDNA from a properly-characterized hPSC line. For calculations of plasmid copies per cell, it’s essential to include internal reference gDNA sequences to allow the quantification of, for example, ribonuclease P (RNaseP) or human telomerase reverse transcriptase (hTERT).
Karyotype-A protracted-time period culture of hESCs can accumulate tradition-pushed mutations [4]. Due to that, it is crucial to pay further consideration to genomic integrity. Karyotype tests might be performed by resuscitating consultant aliquots and culturing them for 48-72 h earlier than harvesting cells for karyotypic analysis. If abnormalities are discovered within the first 20 karyotypes, the evaluation should be repeated on a recent pattern. When this example is repeated, the line is evaluated as abnormal. Repeated abnormalities must be recorded. Although karyology is a crucial procedure in stem cell high quality control, the only nucleotide polymorphism (SNP) array, discussed later, has approximately 50 occasions greater decision.
Viral testing-When assessing the standard of stem cells, all checks for dangerous human adventitious brokers must be performed (e.g. hepatitis C or human immunodeficiency virus). This procedure must be carried out in the case of non-xeno-free culture agents.
Bacteriology-Bacterial or fungal sterility exams might be divided into tradition- or broth-primarily based assessments. All of the procedures must be recommended by pharmacopoeia for the jurisdiction through which the work is performed.
Single nucleotide polymorphism arrays-This procedure is a type of DNA microarray that detects population polymorphisms by enabling the detection of subchromosomal modifications and the copy-impartial loss of heterozygosity, in addition to a sign of cellular transformation. The SNP assay consists of three elements. The primary is labelling fragmented nucleic acid sequences with fluorescent dyes. The second is an array that contains immobilized allele-specific oligonucleotide (ASO) probes. The last part detects, records, and finally interprets the signal.
Flow cytometry-This is a method that makes use of mild to count and profile cells in a heterogeneous fluid mixture. It allows researchers to accurately and quickly accumulate data from heterogeneous fluid mixtures with live cells. Cells are passed through a slim channel one by one. During mild illumination, sensors detect mild emitted or refracted from the cells. The final step is information analysis, compilation and integration into a comprehensive picture of the sample.
Phenotypic pluripotency assays-Recognizing undifferentiated cells is essential in successful stem cell therapy. Among other characteristics, stem cells seem to have a distinct morphology with a excessive nucleus to cytoplasm ratio and a outstanding nucleolus. Cells appear to be flat with defined borders, in distinction to differentiating colonies, which appear as loosely located cells with tough borders [5]. It will be significant that photos of preferrred and poor high quality colonies for every cell line are saved in laboratories, so every time there is doubt about the quality of culture, it may well at all times be checked in response to the representative picture. Embryoid body formation or directed differentiation of monolayer cultures to provide cell types consultant of all three embryonic germ layers must be performed. It can be crucial to notice that colonies cultured under completely different conditions could have completely different morphologies [6].
Histone modification and DNA methylation-Quality management may be achieved by using epigenetic analysis instruments resembling histone modification or DNA methylation. When stem cells differentiate, the methylation course of silences pluripotency genes, which reduces differentiation potential, although different genes might endure demethylation to turn out to be expressed [7]. It is important to emphasize that stem cell identification, together with its morphological characteristics, can be associated to its epigenetic profile [8, 9]. In response to Brindley [10], there’s a relationship between epigenetic adjustments, pluripotency, and cell growth situations, which emphasizes that unmethylated regions appear to be serum-dependent.
hESC derivation and media
hESCs can be derived using a variety of strategies, from traditional culturing to laser-assisted methodologies or microsurgery [11]. hESC differentiation must be specified to keep away from teratoma formation (see Fig. 3).
Spontaneous differentiation of hESCs causes the formation of a heterogeneous cell inhabitants. There is a unique consequence, nonetheless, when commitment signals (in types of soluble components and tradition conditions) are utilized and allow the number of progenitor cells
hESCs spontaneously differentiate into embryonic our bodies (EBs) [12]. EBs could be studied as a substitute of embryos or animals to predict their results on early human improvement. There are many various strategies for buying EBs, reminiscent of bioreactor tradition [13], hanging drop tradition [12], or microwell technology [14, 15]. These methods permit specific precursors to form in vitro [16].
The important half of those culturing procedures is a separation of interior cell mass to tradition future hESCs (Fig. 4) [17]. Rosowski et al. [18] emphasizes that particular consideration must be taken in controlling spontaneous differentiation. When the colony reaches the suitable dimension, cells have to be separated. The occurrence of pluripotent cells lasts for 1-2 days. Because the classical utilization of hESCs brought on moral considerations about gastrulas used during procedures, Chung et al. [19] found out that it’s also potential to obtain hESCs from four cell embryos, leaving the next probability of embryo survival. Additionally, Zhang et al. [20] used solely in vitro fertilization development-arrested cells.
Culturing of pluripotent stem cells in vitro. Three days after fertilization, totipotent cells are formed. Blastocysts with ICM are formed on the sixth day after fertilization. Pluripotent stem cells from ICM can then be successfully transmitted on a dish
Cell passaging is used to type smaller clusters of cells on a new culture surface [21]. There are four necessary passaging procedures.
Enzymatic dissociation is a reducing motion of enzymes on proteins and adhesion domains that bind the colony. It is a gentler method than the guide passage. It’s crucial to not leave hESCs alone after passaging. Solitary cells are extra delicate and might simply endure cell loss of life; collagenase kind IV is an example [22, 23].
Manual passage, on the other hand, focuses on utilizing cell scratchers. The collection of sure cells is just not obligatory. This ought to be completed within the early levels of cell line derivation [24].
Trypsin utilization permits a healthy, automated hESC passage. Good Manufacturing Practice (GMP)-grade recombinant trypsin is broadly accessible in this process [24]. However, there is a risk of reducing the pluripotency and viability of stem cells [25]. Trypsin utilization can be halted with an inhibitor of the protein rho-related protein kinase (ROCK) [26].
Ethylenediaminetetraacetic acid (EDTA) not directly suppresses cell-to-cell connections by chelating divalent cations. Their suppression promotes cell dissociation [27].
Stem cells require a mixture of progress elements and nutrients to differentiate and develop. The medium ought to be modified each day.
Traditional tradition strategies used for hESCs are mouse embryonic fibroblasts (MEFs) as a feeder layer and bovine serum [28] as a medium. Martin et al. [29] demonstrated that hESCs cultured within the presence of animal merchandise specific the non-human sialic acid, N-glycolylneuraminic acid (NeuGc). Feeder layers prevent uncontrolled proliferation with factors comparable to leukaemia inhibitory issue (LIF) [30].
First feeder layer-free tradition can be supplemented with serum substitute, combined with laminin [31]. This causes stable karyotypes of stem cells and pluripotency lasting for over a year.
Initial culturing media may be serum (e.g. foetal calf serum FCS), synthetic replacement reminiscent of synthetic serum substitute (SSS), knockout serum substitute (KOSR), or StemPro [32]. The best tradition medium comprises solely eight essential elements: DMEM/F12 medium, selenium, NaHCO3, l-ascorbic acid, transferrin, insulin, TGFβ1, and FGF2 [33]. It isn’t but fully identified whether culture methods developed for hESCs may be allowed with out adaptation in iPSC cultures.
Turning point in stem cell therapy
The turning point in stem cell therapy appeared in 2006, when scientists Shinya Yamanaka, together with Kazutoshi Takahashi, discovered that it is feasible to reprogram multipotent grownup stem cells to the pluripotent state. This course of avoided endangering the foetus’ life in the method. Retrovirus-mediated transduction of mouse fibroblasts with 4 transcription elements (Oct-3/4, Sox2, KLF4, and c-Myc) [34] which can be primarily expressed in embryonic stem cells may induce the fibroblasts to turn out to be pluripotent (Fig. 5) [35]. This new form of stem cells was named iPSCs. One 12 months later, the experiment additionally succeeded with human cells [36]. After this success, the method opened a new subject in stem cell research with a generation of iPSC lines that can be customized and biocompatible with the affected person. Recently, research have centered on lowering carcinogenesis and enhancing the conduction system.
Retroviral-mediated transduction induces pluripotency in isolated affected person somatic cells. Target cells lose their function as somatic cells and, once again, turn out to be pluripotent and can differentiate into any cell sort of human body
The turning point was influenced by former discoveries that occurred in 1962 and 1987.
The former discovery was about scientist John Gurdon efficiently cloning frogs by transferring a nucleus from a frog’s somatic cells into an oocyte. This prompted a complete reversion of somatic cell improvement [37]. The outcomes of his experiment turned an immense discovery since it was beforehand believed that cell differentiation is a one-means road only, but his experiment instructed the other and demonstrated that it is even attainable for a somatic cell to once more acquire pluripotency [38].
The latter was a discovery made by Davis R.L. that centered on fibroblast DNA subtraction. Three genes were found that originally appeared in myoblasts. The enforced expression of solely one of the genes, named myogenic differentiation 1 (Myod1), triggered the conversion of fibroblasts into myoblasts, displaying that reprogramming cells is possible, and it can even be used to remodel cells from one lineage to another [39].
iPSCs
Although pluripotency can occur naturally only in embryonic stem cells, it is possible to induce terminally differentiated cells to change into pluripotent once more. The process of direct reprogramming converts differentiated somatic cells into iPSC traces that may kind all cell forms of an organism. Reprogramming focuses on the expression of oncogenes resembling Myc and Klf4 (Kruppel-like factor 4). This process is enhanced by a downregulation of genes selling genome stability, corresponding to p53. Additionally, cell reprogramming involves histone alteration. All these processes can cause potential mutagenic danger and later result in an increased number of mutations. Quinlan et al. [40] checked fully pluripotent mouse iPSCs using whole genome DNA sequencing and structural variation (SV) detection algorithms. Based on these research, it was confirmed that though there have been single mutations in the non-genetic region, there have been non-retrotransposon insertions. This led to the conclusion that current reprogramming methods can produce fully pluripotent iPSCs with out extreme genomic alterations.
Throughout the course of growth from pluripotent hESCs to differentiated somatic cells, crucial modifications seem in the epigenetic structure of those cells. There is a restriction or permission of the transcription of genes relevant to each cell kind. When somatic cells are being reprogrammed utilizing transcription factors, all of the epigenetic structure has to be reconditioned to realize iPSCs with pluripotency [41]. However, cells of each tissue endure specific somatic genomic methylation. This influences transcription, which may additional trigger alterations in induced pluripotency [42].
Source of iPSCs
Because pluripotent cells can propagate indefinitely and differentiate into any type of cell, they can be a limiteless source, both for replacing misplaced or diseased tissues. iPSCs bypass the necessity for embryos in stem cell therapy. Because they are made from the patient’s own cells, they are autologous and not generate any threat of immune rejection.
At first, fibroblasts were used as a supply of iPSCs. Because a biopsy was wanted to realize a majority of these cells, the technique underwent further analysis. Researchers investigated whether extra accessible cells might be used in the tactic. Further, other cells had been used in the process: peripheral blood cells, keratinocytes, and renal epithelial cells found in urine. Another strategy to stem cell transplantation can be stimulating a patient’s endogenous stem cells to divide or differentiate, occurring naturally when skin wounds are healing. In 2008, pancreatic exocrine cells had been proven to be reprogrammed to useful, insulin-producing beta cells [43].
The perfect stem cell supply seems to be the fibroblasts, which is extra tempting within the case of logistics since its stimulation may be fast and higher managed [44].
Teratoma formation assay
The self-renewal and differentiation capabilities of iPSCs have gained important curiosity and a focus in regenerative medication sciences. To study their talents, a top quality-management assay is needed, of which one in all the most important is the teratoma formation assay. Teratomas are benign tumours. Teratomas are able to fast development in vivo and are characteristic due to their means to become tissues of all three germ layers simultaneously. Due to the high pluripotency of teratomas, this formation assay is taken into account an evaluation of iPSC’s abilities [45].
Teratoma formation price, as an example, was observed to be elevated in human iPSCs in comparison with that in hESCs [46]. This difference could also be linked to different differentiation methods and cell origins. Most commonly, the teratoma assay includes an injection of examined iPSCs subcutaneously or beneath the testis or kidney capsule in mice, that are immune-deficient [47]. After injection, an immature but recognizable tissue might be observed, such because the kidney tubules, bone, cartilage, or neuroepithelium [30]. The injection site could have an impact on the efficiency of teratoma formation [48].
There are three teams of markers used in this assay to differentiate the cells of germ layers. For endodermal tissue, there is insulin/C-peptide and alpha-1 antitrypsin [49]. For the mesoderm, derivatives can be utilized, e.g. cartilage matrix protein for the bone and alcian blue for the cartilage. As ectodermal markers, class III B botulin or keratin can be used for keratinocytes.
Teratoma formation assays are thought of the gold commonplace for demonstrating the pluripotency of human iPSCs, demonstrating their prospects under physiological conditions. Resulting from their precise tissue formation, they might be used for the characterization of many cell lineages [50].
Directed differentiation
To be useful in therapy, stem cells must be converted into desired cell types as essential or else the entire regenerative drugs course of will likely be pointless. Differentiation of ESCs is crucial because undifferentiated ESCs can cause teratoma formation in vivo. Understanding and using signalling pathways for differentiation is an important methodology in successful regenerative medicine. In directed differentiation, it is likely to mimic indicators which might be obtained by cells when they endure successive phases of improvement [51]. The extracellular microenvironment performs a major function in controlling cell behaviour. By manipulating the tradition situations, it is feasible to restrict particular differentiation pathways and generate cultures which are enriched in sure precursors in vitro. However, attaining a similar effect in vivo is challenging. It is essential to develop tradition conditions that will allow the promotion of homogenous and enhanced differentiation of ESCs into functional and desired tissues.
Regarding the self-renewal of embryonic stem cells, Hwang et al. [52] noted that the best tradition method for hESC-based cell and tissue therapy could be an outlined tradition free of either the feeder layer or animal elements. This is because cell and tissue therapy requires the maintenance of massive portions of undifferentiated hESCs, which does not make feeder cells appropriate for such tasks.
Most directed differentiation protocols are formed to mimic the event of an interior cell mass throughout gastrulation. During this process, pluripotent stem cells differentiate into ectodermal, mesodermal, or endodermal progenitors. Mall molecules or progress elements induce the conversion of stem cells into applicable progenitor cells, which is able to later give rise to the specified cell type. There’s a variety of signal intensities and molecular households which will have an effect on the establishment of germ layers in vivo, comparable to fibroblast progress elements (FGFs) [53]; the Wnt household [54] or superfamily of remodeling development elements-β(TGFβ); and bone morphogenic proteins (BMP) [55]. Each candidate factor have to be examined on numerous concentrations and additionally applied to various durations because the exact concentrations and instances during which developing cells in embryos are influenced during differentiation are unknown. As an illustration, molecular antagonists of endogenous BMP and Wnt signalling can be used for ESC formation of ectoderm [56]. However, transient Wnt and decrease concentrations of the TGFβ household trigger mesodermal differentiation [57]. Regarding endoderm formation, a higher activin A focus could also be required [58, 59].
There are numerous protocols concerning the strategies of forming progenitors of cells of every of germ layers, akin to cardiomyocytes [60], hepatocytes [61], renal cells [62], lung cells [63, 64], motor neurons [65], intestinal cells [66], or chondrocytes [67].
Directed differentiation of either iPSCs or ESCs into, e.g. hepatocytes, might affect and develop the research of the molecular mechanisms in human liver development. As well as, it could additionally present the likelihood to type exogenous hepatocytes for drug toxicity testing [68].
Levels of focus and duration of action with a selected signalling molecule can cause a variety of factors. Unfortunately, for now, a high cost of recombinant components is prone to restrict their use on a larger scale in medicine. The extra promising approach focuses on the use of small molecules. These can be utilized for both activating or deactivating specific signalling pathways. They improve reprogramming efficiency by creating cells that are suitable with the specified kind of tissue. It is a less expensive and non-immunogenic methodology.
One of many successful examples of small-molecule cell therapies is antagonists and agonists of the Hedgehog pathway. They show to be very helpful in motor neuron regeneration [69]. Endogenous small molecules with their operate in embryonic development may also be utilized in in vitro methods to induce the differentiation of cells; for instance, retinoic acid, which is responsible for patterning the nervous system in vivo [70], surprisingly induced retinal cell formation when the laboratory procedure involved hESCs [71].
The efficacy of differentiation factors is determined by useful maturity, efficiency, and, lastly, introducing produced cells to their in vivo equal. Topography, shear stress, and substrate rigidity are components influencing the phenotype of future cells [72].
The management of biophysical and biochemical signals, the biophysical atmosphere, and a correct information of hESC differentiation are important elements in appropriately cultured stem cells.
Stem cell utilization and their manufacturing standards and culture systems
The European Medicines Agency and the Food and Drug Administration have set Good Manufacturing Practice (GMP) pointers for protected and appropriate stem cell transplantation. Prior to now, protocols used for stem cell transplantation required animal-derived products [73].
The danger of introducing animal antigens or pathogens brought about a restriction of their use. Resulting from such limitations, the technique required an apparent update [74]. Now, it is essential to make use of xeno-free equivalents when establishing cell traces which are derived from recent embryos and cultured from human feeder cell traces [75]. On this methodology, it’s essential to change any non-human supplies with xeno-free equivalents [76].
NutriStem with LN-511, TeSR2 with human recombinant laminin (LN-511), and RegES with human foreskin fibroblasts (HFFs) are generally used xeno-free tradition programs [33]. There are various organizations and worldwide initiatives, such because the National Stem Cell Bank, that provide stem cell strains for therapy or medical research [77].
Stem cell use in medication
Stem cells have great potential to change into one in all an important facets of medication. In addition to the fact that they play a big position in creating restorative drugs, their study reveals a lot data in regards to the complicated occasions that happen throughout human improvement.
The distinction between a stem cell and a differentiated cell is mirrored within the cells’ DNA. In the previous cell, DNA is organized loosely with working genes. When alerts enter the cell and the differentiation process begins, genes which might be no longer needed are shut down, however genes required for the specialized perform will remain lively. This process can be reversed, and it is understood that such pluripotency will be achieved by interaction in gene sequences. Takahashi and Yamanaka [78] and Loh et al. [79] discovered that octamer-binding transcription issue 3 and four (Oct3/4), intercourse determining area Y (SRY)-box 2 and Nanog genes operate as core transcription factors in sustaining pluripotency. Among them, Oct3/four and Sox2 are important for the era of iPSCs.
Many serious medical situations, equivalent to delivery defects or cancer, are caused by improper differentiation or cell division. Currently, several stem cell therapies are doable, amongst that are treatments for spinal cord harm, coronary heart failure [80], retinal and macular degeneration [81], tendon ruptures, and diabetes sort 1 [82]. Stem cell research can additional help in better understanding stem cell physiology. This will end result find new methods of treating at the moment incurable diseases.
Haematopoietic stem cell transplantation
Haematopoietic stem cells are essential because they’re by far the most completely characterized tissue-specific stem cell; in spite of everything, they have been experimentally studied for greater than 50 years. These stem cells seem to offer an accurate paradigm model system to study tissue-particular stem cells, and they’ve potential in regenerative drugs.
Multipotent haematopoietic stem cell (HSC) transplantation is at the moment the preferred stem cell therapy. Target cells are normally derived from the bone marrow, peripheral blood, or umbilical cord blood [83]. The procedure could be autologous (when the patient’s personal cells are used), allogenic (when the stem cell comes from a donor), or syngeneic (from an equivalent twin). HSCs are liable for the technology of all functional haematopoietic lineages in blood, including erythrocytes, leukocytes, and platelets. HSC transplantation solves problems which might be brought on by inappropriate functioning of the haematopoietic system, which incorporates diseases corresponding to leukaemia and anaemia. However, when standard sources of HSC are considered, there are some essential limitations. First, there’s a restricted number of transplantable cells, and an efficient approach of gathering them has not yet been found. There can also be a problem with discovering a fitting antigen-matched donor for transplantation, and viral contamination or any immunoreactions also trigger a discount in efficiency in typical HSC transplantations. Haematopoietic transplantation must be reserved for patients with life-threatening diseases because it has a multifactorial character and can be a dangerous process. iPSC use is essential in this procedure. Using a patient’s personal unspecialized somatic cells as stem cells supplies the greatest immunological compatibility and considerably increases the success of the process.
Stem cells as a target for pharmacological testing
Stem cells could be utilized in new drug assessments. Each experiment on dwelling tissue could be performed safely on particular differentiated cells from pluripotent cells. If any undesirable impact seems, drug formulas will be changed until they reach a enough level of effectiveness. The drug can enter the pharmacological market with out harming any stay testers. However, to test the medication properly, the conditions must be equal when evaluating the effects of two drugs. To achieve this aim, researchers need to achieve full management of the differentiation process to generate pure populations of differentiated cells.
Stem cells as a substitute for arthroplasty
One in all the largest fears of professional sportsmen is getting an harm, which most often signifies the tip of their skilled profession. This is applicable particularly to tendon injuries, which, attributable to current therapy choices focusing both on conservative or surgical therapy, typically do not provide acceptable outcomes. Problems with the tendons start with their regeneration capabilities. Instead of functionally regenerating after an injury, tendons merely heal by forming scar tissues that lack the functionality of healthy tissues. Factors which will trigger this failed healing response embrace hypervascularization, deposition of calcific supplies, ache, or swelling [84].
Additionally, in addition to issues with tendons, there is a excessive likelihood of buying a pathological situation of joints known as osteoarthritis (OA) [85]. OA is widespread because of the avascular nature of articular cartilage and its low regenerative capabilities [86]. Although arthroplasty is at present a typical process in treating OA, it isn’t excellent for youthful patients as a result of they will outlive the implant and will require a number of surgical procedures in the future. These are situations the place stem cell therapy can help by stopping the onset of OA [87]. However, these procedures are not properly developed, and the lengthy-time period maintenance of hyaline cartilage requires additional research.
Osteonecrosis of the femoral hip (ONFH) is a refractory illness associated with the collapse of the femoral head and threat of hip arthroplasty in youthful populations [88]. Although total hip arthroplasty (THA) is clinically profitable, it isn’t preferrred for younger patients, largely because of the limited lifetime of the prosthesis. An increasing variety of clinical research have evaluated the therapeutic effect of stem cells on ONFH. Many of the authors demonstrated optimistic outcomes, with decreased pain, improved operate, or avoidance of THA [89,90,91].
Rejuvenation by cell programming
Ageing is a reversible epigenetic course of. The primary cell rejuvenation research was printed in 2011 [92]. Cells from aged individuals have totally different transcriptional signatures, excessive ranges of oxidative stress, dysfunctional mitochondria, and shorter telomeres than in younger cells [93]. There is a speculation that when human or mouse adult somatic cells are reprogrammed to iPSCs, their epigenetic age is nearly reset to zero [94]. This was based mostly on an epigenetic mannequin, which explains that on the time of fertilization, all marks of parenteral ageing are erased from the zygote’s genome and its ageing clock is reset to zero [95].
Of their study, Ocampo et al. [96] used Oct4, Sox2, Klf4, and C-myc genes (OSKM genes) and affected pancreas and skeletal muscle cells, which have poor regenerative capability. Their procedure revealed that these genes can also be used for efficient regenerative treatment [97]. The primary challenge of their technique was the necessity to employ an method that does not use transgenic animals and does not require an indefinitely long utility. The first clinical strategy can be preventive, centered on stopping or slowing the ageing charge. Later, progressive rejuvenation of outdated people will be attempted. Sooner or later, this technique could elevate some moral points, such as overpopulation, resulting in lower availability of food and energy.
For now, it is important to learn how to implement cell reprogramming expertise in non-transgenic elder animals and humans to erase marks of ageing without removing the epigenetic marks of cell identification.
Cell-based mostly therapies
Stem cells may be induced to grow to be a particular cell sort that is required to repair broken or destroyed tissues (Fig. 6). Currently, when the necessity for transplantable tissues and organs outweighs the possible supply, stem cells appear to be an ideal answer for the issue. The commonest circumstances that profit from such therapy are macular degenerations [98], strokes [99], osteoarthritis [89, 90], neurodegenerative diseases, and diabetes [100]. On account of this technique, it will possibly become attainable to generate wholesome coronary heart muscle cells and later transplant them to patients with coronary heart illness.
Stem cell experiments on animals. These experiments are one of the numerous procedures that proved stem cells to be a vital think about future regenerative medicine
In the case of sort 1 diabetes, insulin-producing cells within the pancreas are destroyed on account of an autoimmunological reaction. As an alternative to transplantation therapy, it may be doable to induce stem cells to differentiate into insulin-producing cells [101].
iPS cells with their theoretically limitless propagation and differentiation talents are engaging for the present and future sciences. They can be saved in a tissue financial institution to be a necessary supply of human tissue used for medical examination. The issue with conventional differentiated tissue cells held within the laboratory is that their propagation options diminish after time. This does not happen in iPSCs.
The umbilical cord is thought to be wealthy in mesenchymal stem cells. On account of its cryopreservation instantly after delivery, its stem cells might be efficiently saved and used in therapies to prevent the longer term life-threatening diseases of a given affected person.
Stem cells of human exfoliated deciduous teeth (SHED) present in exfoliated deciduous teeth has the power to become more types of body tissues than different stem cells [102] (Table 1). Techniques of their assortment, isolation, and storage are easy and non-invasive. Among some great benefits of banking, SHED cells are:
– Guaranteed donor-match autologous transplant that causes no immune reaction and rejection of cells [103]
– Simple and painless for each youngster and parent
– Lower than one third of the price of cord blood storage
– Not topic to the same ethical considerations as embryonic stem cells [104]
– In contrast to cord blood stem cells, SHED cells are in a position to regenerate into solid tissues corresponding to connective, neural, dental, or bone tissue [105, 106]
– SHED could be helpful for close kin of the donor
Fertility diseases
In 2011, two researchers, Katsuhiko Hayashi et al. [107], showed in an experiment on mice that it is possible to kind sperm from iPSCs. They succeeded in delivering wholesome and fertile pups in infertile mice. The experiment was also profitable for feminine mice, the place iPSCs formed absolutely practical eggs.
Young adults prone to shedding their spermatogonial stem cells (SSC), mostly most cancers patients, are the main target group that may benefit from testicular tissue cryopreservation and autotransplantation. Effective freezing strategies for adult and pre-pubertal testicular tissue are available [108].
Qiuwan et al. [109] provided vital proof that human amniotic epithelial cell (hAEC) transplantation might successfully improve ovarian function by inhibiting cell apoptosis and lowering inflammation in injured ovarian tissue of mice, and it could possibly be a promising technique for the management of premature ovarian failure or insufficiency in feminine cancer survivors.
For now, reaching profitable infertility remedies in humans seems to be only a matter of time, however there are a number of challenges to beat. First, the method must have high efficiency; second, the possibilities of forming tumours as a substitute of eggs or sperm must be maximally reduced. The last barrier is how you can mature human sperm and eggs within the lab without transplanting them to in vivo circumstances, which may cause either a tumour threat or an invasive procedure.
Therapy for incurable neurodegenerative diseases
Thanks to stem cell therapy, it is feasible not solely to delay the progression of incurable neurodegenerative diseases corresponding to Parkinson’s illness, Alzheimer’s illness (Ad), and Huntington disease, but in addition, most significantly, to take away the source of the problem. In neuroscience, the discovery of neural stem cells (NSCs) has nullified the earlier concept that grownup CNS weren’t able to neurogenesis [110, 111]. Neural stem cells are capable of enhancing cognitive operate in preclinical rodent models of Ad [112,113,114]. Awe et al. [115] clinically derived relevant human iPSCs from pores and skin punch biopsies to develop a neural stem cell-primarily based approach for treating Ad. Neuronal degeneration in Parkinson’s disease (PD) is focal, and dopaminergic neurons can be efficiently generated from hESCs. PD is an ideal disease for iPSC-based mostly cell therapy [116]. However, this therapy remains to be in an experimental part (https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4539501/). Brain tissue from aborted foetuses was used on patients with Parkinson’s disease [117]. Although the outcomes were not uniform, they showed that therapies with pure stem cells are an important and achievable therapy.
Stem cell use in dentistry
Teeth characterize a very difficult material for regenerative drugs. They are difficult to recreate due to their operate in facets equivalent to articulation, mastication, or aesthetics as a result of their complicated structure. Currently, there’s an opportunity for stem cells to turn out to be extra broadly used than artificial materials. Teeth have a large benefit of being the most natural and non-invasive supply of stem cells.
For now, with out the usage of stem cells, the most typical periodontological therapies are either progress components, grafts, or surgical procedure. For example, there are stem cells in periodontal ligament [118, 119], that are able to differentiating into osteoblasts or cementoblasts, and their capabilities had been additionally assessed in neural cells [120]. Tissue engineering is a profitable technique for treating periodontal diseases. Stem cells of the root apical areas are capable of recreate periodontal ligament. One of many attainable strategies of tissue engineering in periodontology is gene therapy performed utilizing adenoviruses-containing progress components [121].
On account of animal research, dentin regeneration is an effective course of that results within the formation of dentin bridges [122].
Enamel is tougher to regenerate than dentin. After the differentiation of ameloblastoma cells into the enamel, the previous is destroyed, and reparation is impossible. Medical studies have succeeded in differentiating bone marrow stem cells into ameloblastoma [123].
Healthy dental tissue has a excessive quantity of normal stem cells, though this quantity is diminished when tissue is either traumatized or inflamed [124]. There are several dental stem cell teams that can be remoted (Fig. 7).
Localization of stem cells in dental tissues. Dental pulp stem cells (DPSCs) and human deciduous teeth stem cells (SHED) are positioned in the dental pulp. Periodontal ligaments stem cells are located within the periodontal ligament. Apical papilla consists of stem cells from the apical papilla (SCAP)
Dental pulp stem cell (DPSC)
These have been the primary dental stem cells isolated from the human dental pulp, which have been [125] positioned inside dental pulp (Table 2). They have osteogenic and chondrogenic potential. Mesenchymal stem cells (MSCs) of the dental pulp, when remoted, appear extremely clonogenic; they are often remoted from adult tissue (e.g. bone marrow, adipose tissue) and foetal (e.g. umbilical cord) [126] tissue, and they’re able to differentiate densely [127]. MSCs differentiate into odontoblast-like cells and osteoblasts to kind dentin and bone. Their best source locations are the third molars [125]. DPSCs are probably the most helpful dental supply of tissue engineering on account of their simple surgical accessibility, cryopreservation risk, increased production of dentin tissues compared to non-dental stem cells, and their anti-inflammatory abilities. These cells have the potential to be a source for maxillofacial and orthopaedic reconstructions or reconstructions even beyond the oral cavity. DPSCs are in a position to generate all buildings of the developed tooth [128]. In particular, beneficial ends in using DPSCs may be achieved when mixed with other new therapies, akin to periodontal tissue photobiomodulation (laser stimulation), which is an environment friendly method in the stimulation of proliferation and differentiation into distinct cell sorts [129]. DPSCs will be induced to type neural cells to help treat neurological deficits.
Stem cells of human exfoliated deciduous teeth (SHED) have a quicker charge of proliferation than DPSCs and differentiate into an even larger variety of cells, e.g. different mesenchymal and non-mesenchymal stem cell derivatives, resembling neural cells [130]. These cells possess one main disadvantage: they kind a non-full dentin/pulp-like complex in vivo. SHED do not endure the identical moral issues as embryonic stem cells. Both DPSCs and SHED are in a position to kind bone-like tissues in vivo [131] and can be used for periodontal, dentin, or pulp regeneration. DPSCs and SHED may be utilized in treating, for example, neural deficits [132]. DPSCs alone have been examined and successfully utilized for alveolar bone and mandible reconstruction [133].
Periodontal ligament stem cells (PDLSCs)
These cells are used in periodontal ligament or cementum tissue regeneration. They will differentiate into mesenchymal cell lineages to produce collagen-forming cells, adipocytes, cementum tissue, Sharpey’s fibres, and osteoblast-like cells in vitro. PDLSCs exist both on the basis and alveolar bone surfaces; however, on the latter, these cells have better differentiation skills than on the previous [134]. PDLSCs have turn into the primary treatment for periodontal regeneration therapy because of their security and efficiency [135, 136].
Stem cells from apical papilla (SCAP)
These cells are mesenchymal structures situated within immature roots. They are remoted from human immature permanent apical papilla. SCAP are the source of odontoblasts and cause apexogenesis. These stem cells could be induced in vitro to type odontoblast-like cells, neuron-like cells, or adipocytes. SCAP have a higher capability of proliferation than DPSCs, which makes them a greater choice for tissue regeneration [137, 138].
Dental follicle stem cells (DFCs)
These cells are free connective tissues surrounding the developing tooth germ. DFCs contain cells that may differentiate into cementoblasts, osteoblasts, and periodontal ligament cells [139, 140]. Additionally, these cells proliferate after even more than 30 passages [141]. DFCs are most commonly extracted from the sac of a 3rd molar. When DFCs are mixed with a treated dentin matrix, they will type a root-like tissue with a pulp-dentin advanced and ultimately kind tooth roots [141]. When DFC sheets are induced by Hertwig’s epithelial root sheath cells, they’ll produce periodontal tissue; thus, DFCs symbolize a very promising materials for tooth regeneration [142].
Pulp regeneration in endodontics
Dental pulp stem cells can differentiate into odontoblasts. There are few strategies that allow the regeneration of the pulp.
The primary is an ex vivo method. Proper stem cells are grown on a scaffold earlier than they’re implanted into the basis channel [143].
The second is an in vivo method. This methodology focuses on injecting stem cells into disinfected root channels after the opening of the in vivo apex. Additionally, using a scaffold is necessary to prevent the motion of cells in the direction of different tissues. For now, solely pulp-like constructions have been created successfully.
Methods of putting stem cells into the basis channel represent are either soft scaffolding [144] or the appliance of stem cells in apexogenesis or apexification. Immature teeth are one of the best supply [145]. Nerve and blood vessel network regeneration are extraordinarily very important to maintain pulp tissue wholesome.
The potential of dental stem cells is mainly concerning the regeneration of damaged dentin and pulp or the restore of any perforations; in the future, it seems to be even attainable to generate the whole tooth. Such an immense success would result in the gradual substitute of implant therapies. Mandibulary and maxillary defects will be one of the complicated dental problems for stem cells to handle.
Acquiring non-dental tissue cells by dental stem cell differentiation
In 2013, it was reported that it is feasible to grow teeth from stem cells obtained extra-orally, e.g. from urine [146]. Pluripotent stem cells derived from human urine had been induced and generated tooth-like buildings. The bodily properties of the constructions had been much like pure ones apart from hardness [127]. Nonetheless, it appears to be a really promising approach as a result of it’s non-invasive and comparatively low-value, and somatic cells can be utilized as an alternative of embryonic cells. More importantly, stem cells derived from urine didn’t form any tumours, and using autologous cells reduces the chances of rejection [147].
Use of graphene in stem cell therapy
Over latest years, graphene and its derivatives have been increasingly used as scaffold supplies to mediate stem cell development and differentiation [148]. Both graphene and graphene oxide (GO) signify excessive in-airplane stiffness [149]. Because graphene has carbon and aromatic community, it really works both covalently or non-covalently with biomolecules; along with its superior mechanical properties, graphene provides versatile chemistry. Graphene exhibits biocompatibility with cells and their correct adhesion. It additionally tested positively for enhancing the proliferation or differentiation of stem cells [148]. After optimistic experiments, graphene revealed great potential as a scaffold and information for particular lineages of stem cell differentiation [150]. Graphene has been efficiently used within the transplantation of hMSCs and their guided differentiation to particular cells. The acceleration expertise of graphene differentiation and division have been also investigated. It was discovered that graphene can serve as a platform with increased adhesion for each progress factors and differentiation chemicals. It was also discovered that π-π binding was accountable for elevated adhesion and performed a crucial function in inducing hMSC differentiation [150].
Therapeutic potential of extracellular vesicle-based mostly therapies
Extracellular vesicles (EVs) can be launched by nearly each cell of an organism, together with stem cells [151], and are concerned in intercellular communication by way of the supply of their mRNAs, lipids, and proteins. As Oh et al. [152] show, stem cells, along with their paracrine elements-exosomes-can develop into potential therapeutics within the remedy of, e.g. skin ageing. Exosomes are small membrane vesicles secreted by most cells (30-120 nm in diameter) [153]. When endosomes fuse with the plasma membrane, they turn out to be exosomes that have messenger RNAs (mRNAs) and microRNAs (miRNAs), some lessons of non-coding RNAs (IncRNAs) and several proteins that originate from the host cell [154]. IncRNAs can bind to particular loci and create epigenetic regulators, which leads to the formation of epigenetic modifications in recipient cells. Due to this function, exosomes are believed to be implicated in cell-to-cell communication and the progression of diseases resembling most cancers [155]. Recently, many studies have also proven the therapeutic use of exosomes derived from stem cells, e.g. pores and skin damage and renal or lung injuries [156].
In skin ageing, an important factor is publicity to UV mild, referred to as “photoageing” [157], which causes extrinsic skin injury, characterized by dryness, roughness, irregular pigmentation, lesions, and skin cancers. In intrinsic pores and skin ageing, on the other hand, the lack of elasticity is a characteristic characteristic. The pores and skin dermis consists of fibroblasts, that are chargeable for the synthesis of crucial skin parts, reminiscent of procollagen or elastic fibres. These components form both fundamental framework extracellular matrix constituents of the pores and skin dermis or play a major role in tissue elasticity. Fibroblast efficiency and abundance lower with ageing [158]. Stem cells can promote the proliferation of dermal fibroblasts by secreting cytokines corresponding to platelet-derived progress factor (PDGF), remodeling growth factor β (TGF-β), and fundamental fibroblast development factor. Huh et al. [159] mentioned that a medium of human amniotic fluid-derived stem cells (hAFSC) positively affected pores and skin regeneration after longwave UV-induced (UVA, 315-four hundred nm) photoageing by rising the proliferation and migration of dermal fibroblasts. It was found that, in addition to the induction of fibroblast physiology, hAFSC transplantation also improved diseases in circumstances of renal pathology, various cancers, or stroke [160, 161].
Oh [162] additionally offered an alternative choice for the therapy of skin wounds, both caused by bodily injury or attributable to diabetic ulcers. Induced pluripotent stem cell-conditioned medium (iPSC-CM) with none animal-derived components induced dermal fibroblast proliferation and migration.
Natural cutaneous wound healing is divided into three steps: haemostasis/inflammation, proliferation, and remodelling. During the crucial step of proliferation, fibroblasts migrate and increase in number, indicating that it is a essential step in skin restore, and components similar to iPSC-CM that affect it will possibly enhance the whole cutaneous wound healing course of. Paracrine actions carried out by iPSCs are additionally necessary for this therapeutic impact [163]. These actions consequence in the secretion of cytokines corresponding to TGF-β, interleukin (IL)-6, IL-8, monocyte chemotactic protein-1 (MCP-1), vascular endothelial development factor (VEGF), platelet-derived growth issue-AA (PDGF-AA), and basic fibroblast progress issue (bFGF). Bae et al. [164] talked about that TGF-β induced the migration of keratinocytes. It was additionally demonstrated that iPSC components can improve pores and skin wound healing in vivo and in vitro when Zhou et al. [165] enhanced wound healing, even after carbon dioxide laser resurfacing in an in vivo research.
Peng et al. [166] investigated the results of EVs derived from hESCs on in vitro cultured retinal glial, progenitor Müller cells, that are identified to differentiate into retinal neurons. EVs seem heterogeneous in measurement and may be internalized by cultured Müller cells, and their proteins are concerned in the induction and maintenance of stem cell pluripotency. These stem cell-derived vesicles have been responsible for the neuronal trans-differentiation of cultured Müller cells uncovered to them. However, the analysis article points out that the procedure was accomplished solely on in vitro acquired retina.
Challenges concerning stem cell therapy
Although stem cells seem like a perfect solution for medicine, there are still many obstacles that have to be overcome in the future. One of the first issues is moral concern.
The commonest pluripotent stem cells are ESCs. Therapies regarding their use firstly had been, and nonetheless are, the source of ethical conflicts. The explanation behind it started when, in 1998, scientists discovered the potential for removing ESCs from human embryos. Stem cell therapy appeared to be very efficient in treating many, even beforehand incurable, diseases. The problem was that when scientists isolated ESCs in the lab, the embryo, which had potential for becoming a human, was destroyed (Fig. 8). Due to this, scientists, seeing a big potential on this remedy method, centered their efforts on making it potential to isolate stem cells without endangering their source-the embryo.
Use of inner cell mass pluripotent stem cells and their stimulation to differentiate into desired cell varieties
For now, whereas hESCs nonetheless stay an ethically debatable source of cells, they are doubtlessly powerful instruments to be used for therapeutic purposes of tissue regeneration. Because of the complexity of stem cell management programs, there is still much to be realized through observations in vitro. For stem cells to change into a well-liked and broadly accessible process, tumour risk have to be assessed. The second problem is to attain successful immunological tolerance between stem cells and the patient’s body. For now, among the best ideas is to use the patient’s personal cells and devolve them into their pluripotent stage of growth.
New cells need to have the flexibility to completely change misplaced or malfunctioning pure cells. Additionally, there is a concern about the opportunity of obtaining stem cells with out the risk of morbidity or ache for both the patient or the donor. Uncontrolled proliferation and differentiation of cells after implementation should also be assessed earlier than its use in a wide number of regenerative procedures on residing patients [167].
One of many arguments that restrict the usage of iPSCs is their notorious role in tumourigenicity. There is a threat that the expression of oncogenes might increase when cells are being reprogrammed. In 2008, a way was found that allowed scientists to take away oncogenes after a cell achieved pluripotency, although it’s not environment friendly yet and takes a longer amount of time. The strategy of reprogramming could also be enhanced by deletion of the tumour suppressor gene p53, but this gene also acts as a key regulator of most cancers, which makes it inconceivable to remove so as to keep away from extra mutations within the reprogrammed cell. The low efficiency of the process is another problem, which is progressively turning into diminished with every year. At first, the speed of somatic cell reprogramming in Yamanaka’s examine was up to 0.1%. Using transcription elements creates a danger of genomic insertion and additional mutation of the goal cell genome. For now, the only ethically acceptable operation is an injection of hESCs into mouse embryos in the case of pluripotency evaluation [168].
Stem cell obstacles sooner or later
Pioneering scientific and medical advances all the time must be carefully policed in order to verify they’re each ethical and secure. Because stem cell therapy already has a large impact on many points of life, it should not be handled otherwise.
Currently, there are several challenges regarding stem cells. First, the most important one is about fully understanding the mechanism by which stem cells perform first in animal fashions. This step can’t be avoided. For the widespread, international acceptance of the process, concern of the unknown is the best problem to beat.
The effectivity of stem cell-directed differentiation should be improved to make stem cells more dependable and trustworthy for a daily patient. The size of the process is another challenge. Future stem cell therapies may be a big obstacle. Transplanting new, absolutely practical organs made by stem cell therapy would require the creation of tens of millions of working and biologically correct cooperating cells. Bringing such complicated procedures into general, widespread regenerative drugs will require interdisciplinary and international collaboration.
The identification and correct isolation of stem cells from a patient’s tissues is another problem. Immunological rejection is a significant barrier to successful stem cell transplantation. With certain sorts of stem cells and procedures, the immune system may acknowledge transplanted cells as foreign our bodies, triggering an immune reaction leading to transplant or cell rejection.
One of the ideas that could make stem cells a “failsafe” is about implementing a self-destruct possibility in the event that they grow to be dangerous. Further growth and versatility of stem cells might cause reduction of therapy prices for folks affected by at the moment incurable diseases. When facing sure organ failure, as a substitute of undergoing extraordinarily expensive drug treatment, the affected person would be able to make the most of stem cell therapy. The impact of a profitable operation can be speedy, and the patient would avoid chronic pharmacological remedy and its inevitable unwanted side effects.
Although these challenges dealing with stem cell science could be overwhelming, the field is making great advances each day. Stem cell therapy is already obtainable for treating several diseases and situations. Their impact on future medicine seems to be important.
After several many years of experiments, stem cell therapy is becoming a magnificent recreation changer for medication. With each experiment, the capabilities of stem cells are rising, though there are nonetheless many obstacles to beat. Regardless, the influence of stem cells in regenerative medication and transplantology is immense. Currently, untreatable neurodegenerative diseases have the opportunity of becoming treatable with stem cell therapy. Induced pluripotency permits the use of a patient’s personal cells. Tissue banks have gotten more and more well-liked, as they gather cells which might be the supply of regenerative medication in a battle against current and future diseases. With stem cell therapy and all its regenerative benefits, we are better in a position to prolong human life than at any time in historical past.
Abbreviations
Basic fibroblast growth factor
Bone morphogenic proteins
Dental follicle stem cells
Dental pulp stem cells
Embryonic bodies
Fibroblast progress factors
Good Manufacturing Practice
Graphene oxide
Human amniotic fluid-derived stem cells
Human embryonic stem cells
Human foreskin fibroblasts
Inner cell mass
Non-coding RNA
Induced pluripotent stem cells
In vitro fertilization
Knockout serum alternative
Leukaemia inhibitory issue
Monocyte chemotactic protein-1
Fibroblasts
Messenger RNA
Mesenchymal stem cells of dental pulp
Myogenic differentiation
Osteoarthritis
Octamer-binding transcription factor 3 and four
Platelet-derived progress issue
Platelet-derived growth issue-AA
Periodontal ligament stem cells
Rho-related protein kinase
Stem cells from apical papilla
Stem cells of human exfoliated deciduous teeth
Synthetic Serum Substitute
Trophectoderm
Vascular endothelial progress issue
Transforming development elements
Sukoyan MA, Vatolin SY, et al. Embryonic stem cells derived from morulae, interior cell mass, and blastocysts of mink: comparisons of their pluripotencies. Embryo Dev. 1993;36(2):148-58
Larijani B, Esfahani EN, Amini P, Nikbin B, Alimoghaddam K, Amiri S, Malekzadeh R, Yazdi NM, Ghodsi M, Dowlati Y, Sahraian MA, Ghavamzadeh A. Stem cell therapy in remedy of different diseases. Acta Medica Iranica. 2012:79-96 https://www.ncbi.nlm.nih.gov/pubmed/22359076.
Sullivan S, Stacey GN, Akazawa C, et al. Quality tips for clinical-grade human induced pluripotent stem cell traces. Regenerative Med. 2018; https://doi.org/10.2217/rme-2018-0095.
Amps K, Andrews PW, et al. Screening ethnically various human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring progress advantage. Nat. Biotechnol. 2011;29(12):1121-44.
Amit M, Itskovitz-Eldor J. Atlas of human pluripotent stem cells: derivation and culturing. New York: Humana Press; 2012.
Ludwig TE, Bergendahl V, Levenstein ME, Yu J, Probasco MD, Thomson JA. Feeder-independent tradition of human embryonic stem cells. Nat Methods. 2006;3:637-46.
Kang MI. Transitional CpG methylation between promoters and retroelements of tissue-particular genes during human mesenchymal cell differentiation. J. Cell Biochem. 2007;102:224-39.
Vaes B, Craeye D, Pinxteren J. Quality management throughout manufacture of a stem cell therapeutic. BioProcess Int. 2012;10:50-5.
Bloushtain-Qimron N. Epigenetic patterns of embryonic and adult stem cells. Cell Cycle. 2009;8:809-17.
Brindley DA. Peak serum: implications of serum provide for cell therapy manufacturing. Regenerative Medicine. 2012;7:809-17.
Solter D, Knowles BB. Immunosurgery of mouse blastocyst. Proc Natl Acad Sci U S A. 1975;72:5099-102.
Hoepfl G, Gassmann M, Desbaillets I. Differentiating embryonic stem cells into embryoid our bodies. Methods Mole Biol. 2004;254:79-98 https://doi.org/10.1385/1-59259-741-6:079.
Lim WF, Inoue-Yokoo T, Tan KS, Lai MI, Sugiyama D. Hematopoietic cell differentiation from embryonic and induced pluripotent stem cells. Stem Cell Res Ther. 2013;4(3):71. https://doi.org/10.1186/scrt222.
Mohr JC, de Pablo JJ, Palecek SP. 3-D microwell culture of human embryonic stem cells. Biomaterials. 2006;27(36):6032-42. https://doi.org/10.1016/j.biomaterials.2006.07.012.
Doetschman TC, Eistetter H, Katz M, Schmidt W, Kemler R. The in vitro improvement of blastocyst-derived embryonic stem cell strains: formation of the visceral yolk sac, blood islands, and myocardium. J Embryol Exp Morphol. 1985;87:27-45.
Kurosawa HY. Methods for inducing embryoid physique formation: in vitro differentiation system of embryonic stem cells. J Biosci Bioeng. 2007;103:389-98.
Heins N, Englund MC, Sjoblom C, Dahl U, Tonning A, Bergh C, Lindahl A, Hanson C, Semb H. Derivation, characterization, and differentiation of human embryonic stem cells. Stem Cells. 2004;22:367-76.
Rosowski KA, Mertz AF, Norcross S, Dufresne ER, Horsley V. Edges of human embryonic stem cell colonies display distinct mechanical properties and differentiation potential. Sci Rep. 2015;5:Article quantity:14218.
Chung Y, Klimanskaya I, Becker S, Li T, Maserati M, Lu SJ, Zdravkovic T, Ilic D, Genbacev O, Fisher S, Krtolica A, Lanza R. Human embryonic stem cell strains generated without embryo destruction. Cell Stem Cell. 2008;2:113-7.
Zhang X, Stojkovic P, Przyborski S, Cooke M, Armstrong L, Lako M, Stojkovic M. Derivation of human embryonic stem cells from creating and arrested embryos. Stem Cells. 2006;24:2669-76.
Beers J, Gulbranson DR, George N, Siniscalchi LI, Jones J, Thomson JA, Chen G. Passaging and colony expansion of human pluripotent stem cells by enzyme-free dissociation in chemically outlined tradition situations. Nat Protoc. 2012;7:2029-40.
Ellerström C, Hyllner J, Strehl R. single cell enzymatic dissociation of human embryonic stem cells: a easy, robust, and standardized tradition technique. In: Turksen K, editor. Human embryonic stem cell protocols. Methods in molecular biology: Humana Press; 2009. p. 584.
Heng BC, Liu H, Ge Z, Cao T. Mechanical dissociation of human embryonic stem cell colonies by guide scraping after collagenase remedy is way more detrimental to cellular viability than is trypsinization with gentle pipetting. Biotechnol Appl Biochem. 2010;47(1):33-7.
Ellerstrom C, Strehl R, Noaksson K, Hyllner J, Semb H. Facilitated growth of human embryonic stem cells by single-cell enzymatic dissociation. Stem Cells. 2007;25:1690-6.
Brimble SN, Zeng X, Weiler DA, Luo Y, Liu Y, Lyons IG, Freed WJ, Robins AJ, Rao MS, Schulz TC. Karyotypic stability, genotyping, differentiation, feeder-free upkeep, and gene expression sampling in three human embryonic stem cell lines deri. Stem Cells Dev. 2004;13:585-97.
Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T, Takahashi JB, Nishikawa S, Nishikawa S, Muguruma K, Sasai Y. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007;25:681-6.
Nie Y, Walsh P, Clarke DL, Rowley JA, Fellner T. Scalable passaging of adherent human pluripotent stem cells. 2014. https://doi.org/10.1371/journal.pone.0088012.
Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell strains derived from human blastocysts. Science. 1998;282:1145-7.
Martin MJ, Muotri A, Gage F, Varki A. Human embryonic stem cellsexpress an immunogenic nonhuman sialic acid. Nat. Med. 2005;11:228-32.
Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl M, Rogers D. Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature. 1988;336(6200):688-90. https://doi.org/10.1038/336688a0.
Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK. Feeder-free development of undifferentiated human embryonic stem cells. Nature Biotechnol. 2001;19:971-4. https://doi.org/10.1038/nbt1001-971.
Article CAS Google Scholar
Weathersbee PS, Pool TB, Ord T. Synthetic serum substitute (SSS): a globulin-enriched protein complement for human embryo culture. J. Assist Reprod Genet. 1995;12:354-60.
Chen G, Gulbranson DR, Hou Z, Bolin JM, Ruotti V, Probasco MD, Smuga-Otto K, Howden SE, Diol NR, Propson NE, Wagner R, Lee GO, Antosiewicz-Bourget J, Teng JM, Thomson JA. Chemically outlined situations for human iPSC derivation and tradition. Nat. Methods. 2011;8:424-9.
Sommer CA, Mostoslavsky G. Experimental approaches for the generation of induced pluripotent stem cells. Stem Cell Res Ther. 2010;1:26.
Takahashi K, Yamanaka S. Induced pluripotent stem cells in medication and biology. Development. 2013;140(12):2457-sixty one https://doi.org/10.1242/dev.092551.
Shi D, Lu F, Wei Y, et al. Buffalos (Bubalus bubalis) cloned by nuclear transfer of somatic cells. Biol. Reprod. 2007;77:285-91. https://doi.org/10.1095/biolreprod.107.060210.
Gurdon JB. The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. Development. 1962;10:622-40 http://dev.biologists.org/content/10/4/622.
Kain K. The start of cloning: an interview with John Gurdon. Dis Model Mech. 2009;2(1-2):9-10. https://doi.org/10.1242/dmm.002014.
Article PubMed Central Google Scholar
Davis RL, Weintraub H, Lassar AB. Expression of a single transfected cDNA converts fibroblasts to myoblasts. Cell. 1987;24(51(6)):987-1000.
Quinlan AR, Boland MJ, Leibowitz ML, et al. Genome sequencing of mouse induced pluripotent stem cells reveals retroelement stability and infrequent DNA rearrangement throughout reprogramming. Cell Stem Cell. 2011;9(4):366-73.
Maherali N, Sridharan R, Xie W, Utika LJ, Eminli S, Arnold K, Stadtfeld M, Yachechko R, Tchieu J, Jaenisch R, Plath K, Hochedlinger K. Directly reprogrammed fibroblasts present global epigenetic remodeling and widespread tissue contribution. Cell Stem Cell. 2007;1:55-70.
Ohi Y, Qin H, Hong C, Blouin L, Polo JM, Guo T, Qi Z, Downey SL, Manos PD, Rossi DJ, Yu J, Hebrok M, Hochedlinger K, Costello JF, Song JS, Ramalho-Santos M. Incomplete DNA methylation underlines a transcriptional reminiscence of somatic cells in human IPS cells. Nat Cell Biol. 2011;13:541-9.
Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA. In vivo reprogramming of adult pancreatic exocrine cells to beta-cells. Nature. 2008;455:627-32 https://doi.org/10.1038/nature07314.
Hilfiker A, Kasper C, Hass R, Haverich A. Mesenchymal stem cells and progenitor cells in connective tissue engineering and regenerative medication: is there a future for transplantation? Langenbecks Arch Surg. 2011;396:489-97.
Zhang Wendy, Y., de Almeida Patricia, E., and Wu Joseph, C. Teratoma formation: a device for monitoring pluripotency in stem cell research. StemBook, ed. The Stem Cell Research Community. June 12, 2012. https://doi.org/10.3824/stembook.1.53.1.
Narsinh KH, Sun N, Sanchez-Freire V, et al. Single cell transcriptional profiling reveals heterogeneity of human induced pluripotent stem cells. J Clin Invest. 2011;121(3):1217-21.
Gertow K, Przyborski S, Loring JF, Auerbach JM, Epifano O, Otonkoski T, Damjanov I, AhrlundRichter L. Isolation of human embryonic stem cell-derived teratomas for the evaluation of pluripotency. Curr Protoc Stem Cell Biol. 2007, Chapter 1, Unit 1B 4. 3: 1B.4.1-1B.4.29.
Cooke MJ, Stojkovic M, Przyborski SA. Growth of teratomas derived from human pluripotent stem cells is influenced by the graft site. Stem Cells Dev. 2006;15(2):254-9.
Przyborski SA. Differentiation of human embryonic stem cells after transplantation in immune-deficient mice. Stem Cells. 2005;23:1242-50.
Tannenbaum SE, Turetsky TT, Singer O, Aizenman E, Kirshberg S, Ilouz N, Gil Y, Berman-Zaken Y, Perlman TS, Geva N, Levy O, Arbell D, Simon A, Ben-Meir A, Shufaro Y, Laufer N, Reubinoff BE. Derivation of xeno-free and GMP-grade human embryonic stem cells- platforms for future clinical purposes. PLoS One. 2012;7:e35325.
Cohen DE, Melton D. Turning straw into gold: directing cell fate for regenerative medicine. Nat Rev Genet. 2011;12:243-52.
Hwang NS, Varghese S, Elisseeff J. Controlled differentiation of stem cells. Adv Drug Deliv Rev. 2007;60(2):199-214. https://doi.org/10.1016/j.addr.2007.08.036.
Turner N, Grose R. Fibroblast development issue signalling: from improvement to most cancers. Nat Rev Cancer. 2010;10:116-29.
Rao TP, Kuhl M. An up to date overview on Wnt signaling pathways: a prelude for more. Circ Res. 2010;106:1798-806.
Moustakas A, Heldin CH. The regulation of TGFbeta sign transduction. Development. 2009;136:3699-714.
Efthymiou AG, Chen G, Rao M, Chen G, Boehm M. Self-renewal and cell lineage differentiation methods in human embryonic stem cells and induced pluripotent stem cells. Expert Opin Biol Ther. 2014;14:1333-44.
Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ, Kennedy M, Henckaerts E, Bonham K, Abbott GW, Linden RM, Field LJ, Keller GM. Human cardiovascular progenitor cells develop from a KDRþembryonic-stem-cell-derived inhabitants. Nature. 2008;453:524-8.
Kroon E, Martinson LA, Kadoya K, Bang AG, Kelly OG, Eliazer S, Young H, Richardson M, Smart NG, Cunningham J, Agulnick Ad, D’amour KA, Carpenter MK, Baetge EE. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008;26(4):443-52. https://doi.org/10.1038/nbt1393.
Vallier L, Reynolds D, Pedersen RA. Nodal inhibits differentiation of human embryonic stem cells alongside the neuroectodermal default pathway. Dev Biol. 2004;275:403-21.
Burridge PW, Zambidis ET. Highly environment friendly directed differentiation of human induced pluripotent stem cells into cardiomyocytes. Methods Mol Biol. 2013;997:149-61.
Cai J, Zhao Y, Liu Y, Ye F, Song Z, Qin H, Meng S, Chen Y, Zhou R, Song X, Guo Y, Ding M, Deng H. Directed differentiation of human embryonic stem cells into practical hepatic cells. Hepatology. 2007;45:1229-39.
Takasato M, Er PX, Becroft M, Vanslambrouck JM, Stanley EG, Elefanty AG, Little MH. Directing human embryonic stem cell differentiation in direction of a renal lineage generates a selforganizing kidney. Nat Cell Biol. 2014;16:118-26.
Huang SX, Islam MN, O’Neill J, Hu Z, Yang YG, Chen YW, Mumau M, Green MD, VunjakNovakovic G, Bhattacharya J, Snoeck HW. Efficient era of lung and airway epithelial cells from human pluripotent stem cells. Nat Biotechnol. 2014;32:84-91.
Kadzik RS, Morrisey EE. Directing lung endoderm differentiation in pluripotent stem cells. Cell Stem Cell. 2012;10:355-61.
Wichterle H, Lieberam I, Porter JA, Jessell TM. Directed differentiation of embryonic stem cells into motor neurons. Cell. 2002;110:385-97.
Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, Shroyer NF, Wells JM. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105-9.
Oldershaw RA, Baxter MA, Lowe ET, Bates N, Grady LM, Soncin F, Brison DR, Hardingham TE, Kimber SJ. Directed differentiation of human embryonic stem cells towards chondrocytes. Nat Biotechnol. 2010;28:1187-94.
Jun Cai, Ann DeLaForest, Joseph Fisher, Amanda Urick, Thomas Wagner, Kirk Twaroski, Max Cayo, Masato Nagaoka, Stephen A Duncan. Protocol for directed differentiation of human pluripotent stem cells towards a hepatocyte destiny. 2012. DOI: https://doi.org/10.3824/stembook.1.52.1.
Frank-Kamenetsky M, Zhang XM, Bottega S, Guicherit O, Wichterle H, Dudek H, Bumcrot D, Wang FY, Jones S, Shulok J, Rubin LL, Porter JA. Small-molecule modulators of hedgehog signaling: identification and characterization of smoothened agonists and antagonists. J Biol. 2002;1:10.
Oshima K, Shin K, Diensthuber M, Peng AW, Ricci AJ, Heller S. Mechanosensitive hair celllike cells from embryonic and induced pluripotent stem cells. Cell. 2010;141:704-16.
Osakada F, Jin ZB, Hirami Y, Ikeda H, Danjyo T, Watanabe K, Sasai Y, Takahashi M. In vitro differentiation of retinal cells from human pluripotent stem cells by small-molecule induction. J Cell Sci. 2009;122:3169-79.
Kshitiz PJ, Kim P, Helen W, Engler AJ, Levchenko A, Kim DH. Control of stem cell destiny and function by engineering physical microenvironments. Intergr Biol (Camb). 2012;4:1008-18.
Amps K, Andrews PW, Anyfantis G, Armstrong L, Avery S, Baharvand H, Baker J, Baker D, Munoz MB, Beil S, Benvenisty N, Ben-Yosef D, Biancotti JC, Bosman A, Brena RM, Brison D, Caisander G, Camarasa MV, Chen J, ChiaoE CYM, Choo AB, Collins D, et al. Screening ethnically diverse human embryonic stem cells identifies a chromosome 20 minimal amplicon conferring progress advantage. Nat Biotechnol. 2011;29:1132-44.
Nukaya D, Minami K, Hoshikawa R, Yokoi N, Seino S. Preferential gene expression and epigenetic memory of induced pluripotent stem cells derived from mouse pancreas. Genes Cells. 2015;20:367-81.
Murdoch A, Braude P, Courtney A, Brison D, Hunt C, Lawford-Davies J, Moore H, Stacey G, Sethe S, Procurement Working Group Of National Clinical H, E. S. C. F, National Clinical H, E. S. C. F. The procurement of cells for the derivation of human embryonic stem cell strains for therapeutic use: suggestions for good practice. Stem Cell Rev. 2012;8:91-9.
Hewitson H, Wood V, Kadeva N, Cornwell G, Codognotto S, Stephenson E, Ilic D. Generation of KCL035 analysis grade human embryonic stem cell line carrying a mutation in HBB gene. Stem Cell Res. 2016;16:210-2.
Daley GQ, Hyun I, Apperley JF, Barker RA, Benvenisty N, Bredenoord AL, Breuer CK, Caulfield T, Cedars MI, Frey-Vasconcells J, Heslop HE, Jin Y, Lee RT, Mccabe C, Munsie M, Murry CE, Piantadosi S, Rao M, Rooke HM, Sipp D, Studer L, Sugarman J, et al. Setting world requirements for stem cell research and clinical translation: the 2016 ISSCR tips. Stem Cell Rep. 2016;6:787-97.
Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-76. https://doi.org/10.1016/j.cell.2006.07.024.
Loh YH, Wu Q, Chew JL, Vega VB, Zhang W, Chen X, Bourque G, George J, Leong B, Liu J, et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nat Genet. 2006;38:431-40.
Menasche P, Vanneaux V, Hagege A, Bel A, Cholley B, Cacciapuoti I, Parouchev A, Benhamouda N, Tachdjian G, Tosca L, Trouvin JH, Fabreguettes JR, Bellamy V, Guillemain R, SuberbielleBoissel C, Tartour E, Desnos M, Larghero J. Human embryonic stem cell-derived cardiac progenitors for extreme coronary heart failure remedy: first clinical case report. Eur Heart J. 2015;36:2011-7.
Schwartz SD, Regillo CD, Lam BL, Eliott D, Rosenfeld PJ, Gregori NZ, Hubschman JP, Davis JL, Heilwell G, Spirn M, Maguire J, Gay R, Bateman J, Ostrick RM, Morris D, Vincent M, Anglade E, Del Priore LV, Lanza R. Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt’s macular dystrophy: observe-up of two open-label phase 1/2 studies. Lancet. 2015;385:509-16.
Ilic D, Ogilvie C. Concise review: human embryonic stem cells-what have we completed? What are we doing? Where are we going? Stem Cells. 2017;35:17-25.
Rocha V, et al. Clinical use of umbilical cord blood hematopoietic stem cells. Biol Blood Marrow Transplant. 2006;12(1):34-4.
Longo UG, Ronga M, Maffulli N. Sports Med Arthrosc 17:112-126. Achilles tendinopathy. Sports Med Arthrosc. 2009;17:112-26.
Tempfer H, Lehner C, Grütz M, Gehwolf R, Traweger A. Biological augmentation for tendon repair: classes to be discovered from improvement, disease, and tendon stem cell research. In: Gimble J, Marolt D, Oreffo R, Redl H, Wolbank S, editors. Cell engineering and regeneration. Reference Series in Biomedical Engineering. Cham: Springer; 2017.
Goldring MB, Goldring SR. Osteoarthritis. J Cell Physiol. 2007;213:626-34.
Widuchowski W, Widuchowski J, Trzaska T. Articular cartilage defects: examine of 25,124 knee arthroscopies. Knee. 2007;14:177-82.
Li R, Lin Q-X, Liang X-Z, Liu G-B, et al. Stem cell therapy for treating osteonecrosis of the femoral head: from clinical functions to associated basic research. Stem Cell Res Therapy. 2018;9:291 https://doi.org/10.1186/s13287-018-1018-7.
Gangji V, De Maertelaer V, Hauzeur JP. Autologous bone marrow cell implantation within the therapy of non-traumatic osteonecrosis of the femoral head: 5 year observe-up of a prospective managed study. Bone. 2011;49(5):1005-9.
Zhao D, Cui D, Wang B, Tian F, Guo L, Yang L, et al. Treatment of early stage osteonecrosis of the femoral head with autologous implantation of bone marrow-derived and cultured mesenchymal stem cells. Bone. 2012;50(1):325-30.
Sen RK, Tripathy SK, Aggarwal S, Marwaha N, Sharma RR, Khandelwal N. Early results of core decompression and autologous bone marrow mononuclear cells instillation in femoral head osteonecrosis: a randomized management study. J Arthroplast. 2012;27(5):679-86.
Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, Aït-Hamou N, Leschik J, Pellestor F, Ramirez JM, De Vos J, Lehmann S, Lemaitre JM. Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state. Genes Dev. 2011;25:2248-53.
Sahin E, Depinho RA. Linking practical decline of telomeres, mitochondria and stem cells during ageing. Nature. 2010;464:520-8.
Petkovich DA, Podolskiy DI, Lobanov AV, Lee SG, Miller RA, Gladyshev VN. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 2017;25:954-60 https://doi.org/10.1016/j.cmet.2017.03.016.
Gerontology, Rejuvenation by cell reprogramming: a new horizon in. Rodolfo G. Goya, Marianne Lehmann, Priscila Chiavellini, Martina Canatelli-Mallat, Claudia B. Hereñú and Oscar A. Brown. Stem Cell Res Therapy. 2018, 9:349. https://doi.org/10.1186/s13287-018-1075-y.
Ocampo A, Reddy P, Martinez-Redondo P, Platero-Luengo A, Hatanaka F, Hishida T, Li M, Lam D, Kurita M, Beyret E, Araoka T, Vazquez-Ferrer E, Donoso D, Roman JLXJ, Rodriguez-Esteban C, Nuñez G, Nuñez Delicado E, Campistol JM, Guillen I, Guillen P, Izpisua. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell. 2016;167:1719-33.
de Lázaro I, Cossu G, Kostarelos K. Transient transcription factor (OSKM) expression is key towards clinical translation of in vivo cell reprogramming. EMBO Mol Med. 2017;9:733-6.
Sun S, Li ZQ, Glencer P, Cai BC, Zhang XM, Yang J, Li XR. Bringing the age-associated macular degeneration excessive-danger allele age-related maculopathy susceptibility 2 into focus with stem cell expertise. Stem Cell Res Ther. 2017;8:135 https://doi.org/10.1186/s13287-017-0584-4.
Liu J. Induced pluripotent stem cell-derived neural stem cells: new hope for stroke? Stem Cell Res Ther. 2013;4:A hundred and fifteen https://doi.org/10.1186/scrt326.
Shahjalal HM, Dayem AA, Lim KM, Jeon TI, Cho SG. Generation of pancreatic β cells for therapy of diabetes: advances and challenges. Stem Cell ResTher. 2018;9:355 https://doi.org/10.1186/s13287-018-1099-3.
Kroon E, Martinson LA, et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat Biotechnol. 2008;26;443-52.
Arora V, Pooja A, Munshi AK. Banking stem cells from human exfoliated deciduous teeth. J Clin Pediatr Dent. 2009;33(4):289-94.
Mao JJ. Stem cells and the way forward for dental care. New York State Dental J. 2008;74(2):21-4.
Reznick, Jay B. Continuing training: stem cells: rising medical and dental therapies for the dental Professional. Dentaltown Magazine. 2008, pp. 42-53.
Arthur A, Rychkov G, Shi S, Koblar SA, Gronthos S. Adult human dental pulp stem cells differentiate toward functionally active neurons beneath appropriate environmental cues. Stem Cells. 2008;26(7):1787-95.
Cordeiro MM, Dong Z, Kaneko T, Zhang Z, Miyazawa M, Shi S, Smith A. Dental pulp tissue engineering with stem cells from exfoliated. J Endod. 2008;34(8):962-9.
Hayashi K, Ohta H, Kurimoto K, Aramaki S, Saitou M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell. 2011;146(4):519-32. https://doi.org/10.1016/j.cell.2011.06.052.
Sadri-Ardekani H, Atala A. Testicular tissue cryopreservation and spermatogonial stem cell transplantation to revive fertility: from bench to bedside. Stem Cell ResTher. 2014;5:68 https://doi.org/10.1186/scrt457.
Zhang Q, Xu M, Yao X, Li T, Wang Q, Lai D. Human amniotic epithelial cells inhibit granulosa cell apoptosis induced by chemotherapy and restore the fertility. Stem Cell Res Ther. 2015;6:152 https://doi.org/10.1186/s13287-015-0148-4.
Ma DK, Bonaguidi MA, Ming GL, Song H. Adult neural stem cells in the mammalian central nervous system. Cell Res. 2009;19:672-82. https://doi.org/10.1038/cr.2009.56.
Dantuma E, Merchant S, Sugaya K. Stem cells for the treatment of neurodegenerative diseases. Stem Cell ResTher. 2010;1:37 https://doi.org/10.1186/scrt37.
Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A, Kawanishi M, Kawai N, Tamiya T, Nagao S. Neural stem cells transplantation in cortex in a mouse model of Alzheimer’s illness. J Med Invest. 2006;53:61-9. https://doi.org/10.2152/jmi.53.61.
Moghadam FH, Alaie H, Karbalaie K, Tanhaei S, Nasr Esfahani MH, Baharvand H. Transplantation of primed or unprimed mouse embryonic stem cell-derived neural precursor cells improves cognitive operate in Alzheimerian rats. Differentiation. 2009;78:59-68. https://doi.org/10.1016/j.diff.2009.06.005.
Byrne JA. Developing neural stem cell-primarily based remedies for neurodegenerative diseases. Stem Cell ResTher. 2014;5:72. https://doi.org/10.1186/scrt461.
Awe JP, Lee Pc, Ramathal C, Vega-Crespo A, Durruthy-Durruthy J, Cooper A, Karumbayaram S, Lowry WE, Clark AT, Zack JA, Sebastiano V, Kohn DB, Pyle Ad, Martin MG, Lipshutz GS, Phelps PE, Pera RA, Byrne JA. Generation and characterization of transgene-free human induced pluripotent stem cells and conversion to putative clinical-grade status. Stem Cell Res Ther. 2013;4:87. https://doi.org/10.1186/scrt246.
Peng J, Zeng X. The role of induced pluripotent stem cells in regenerative medicine: neurodegenerative diseases. Stem Cell ResTher. 2011;2:32. https://doi.org/10.1186/scrt73.
Wright BL, Barker RA. Established and rising therapies for Huntington’s illness. 2007;7(6):579-87 https://www.ncbi.nlm.nih.gov/pubmed/17896994/579-87.
Lin NH, Gronthos S, Bartold PM. Stem cells and periodontal regeneration. Aust Dent J. 2008;53:108-21.
Seo BM, Miura M, Gronthos S, Bartold PM, Batouli S, Brahim J, et al. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet. 2004;364:149-55.
Ramseier CA, Abramson ZR, Jin Q, Giannobile WV. Gene therapeutics for periodontal regenerative medication. Dent Clin North Am. 2006;50:245-63.
Shi S, Bartold PM, Miura M, Seo BM, Robey PG, Gronthos S. The efficacy of mesenchymal stem cells to regenerate and restore dental structures. OrthodCraniofac Res. 2005;8:191-9.
Iohara K, Nakashima M, Ito M, Ishikawa M, Nakasima A, Akamine A. Dentin regeneration by dental pulp stem cell therapy with recombinant human bone morphogenetic protein. J Dent Res. 2004;83:590-5.
Hu B, Unda F, Bopp-Kuchler S, Jimenez L, Wang XJ, Haikel Y, et al. Bone marrow cells can give rise to ameloblast-like cells. J Dent Res. 2006;85:416-21.
Liu Y, Liu W, Hu C, Xue Z, Wang G, Ding B, Luo H, Tang L, Kong X, Chen X, Liu N, Ding Y, Jin Y. MiR-17 modulates osteogenic differentiation by a coherent feed-ahead loop in mesenchymal stem cells isolated from periodontal ligaments of patients with periodontitis. Stem Cells. 2011;29(11):1804-16. https://doi.org/10.1002/stem.728.
Raspini G, Wolff J, Helminen M, Raspini G, Raspini M, Sándor GK. Dental stem cells harvested from third molars mixed with bioactive glass can induce signs of bone formation in vitro. J Oral Maxillofac Res. 2018;9(1):e2. Published 2018 Mar 31. https://doi.org/10.5037/jomr.2018.9102.
Christodoulou I, Goulielmaki M, Devetzi M, Panagiotidis M, Koliakos G, Zoumpourlis V. Mesenchymal stem cells in preclinical most cancers cytotherapy: a scientific evaluate. Stem Cell Res Ther. 2018;9(1;336). https://doi.org/10.1186/s13287-018-1078-8.
Bansal R, Jain A. Current overview on dental stem cells functions in regenerative dentistry. J Nat Sci Biol Med. 2015;6(1):29-34. https://doi.org/10.4103/0976-9668.149074.
Edgar Ledesma-Martínez, Víctor Manuel Mendoza-Núñez, Edelmiro Santiago-Osorio. Mesenchymal stem cells derived from dental pulp: a overview. Stem Cells Int. 2016, 4,709,572, p. doi: https://doi.org/10.1155/2016/4709572].
Grzech-Leśniak K. Making use of lasers in periodontal treatment: a brand new gold commonplace? Photomed Laser Surg. 2017;35(10):513-4.
Miura M, Gronthos S, Zhao M, Lu B, Fisher LW, Robey PG, Shi S. SHED: stem cells from human exfoliated deciduous teeth. Proc Natl Acad Sci U S A. 2003;100(10):5807-12. https://doi.org/10.1073/pnas.0937635100.
Yasui T, Mabuchi Y, Toriumi H, Ebine T, Niibe K, Houlihan DD, Morikawa S, Onizawa K, Kawana H, Akazawa C, Suzuki N, Nakagawa T, Okano H, Matsuzaki Y. Purified human dental pulp stem cells promote osteogenic regeneration. J Dent Res. 2016;95(2):206-14. https://doi.org/10.1177/0022034515610748.
Yamamoto A, Sakai K, Matsubara K, Kano F, Ueda M. Multifaceted neuro-regenerative actions of human dental pulp stem cells for practical recovery after spinal cord injury. Neurosci Res. 2014;78:16-20. https://doi.org/10.1016/j.neures.2013.10.010.
Article PubMed Google Scholar
d’Aquino R, De Rosa A, Lanza V, Tirino V, Laino L, Graziano A, Desiderio V, Laino G, Papaccio G. Human mandible bone defect restore by the grafting of dental pulp stem/progenitor cells and collagen sponge biocomplexes. Eur Cell Mater. 2009;12, PMID: 19908196:75-83.
Wang L, Shen H, Zheng W, Tang L, Yang Z, Gao Y, Yang Q, Wang C, Duan Y, Jin Y. Characterization of stem cells from alveolar periodontal ligament. Tissue Eng. Part A. 2011;17(7-8):1015-26. https://doi.org/10.1089/ten.tea.2010.0140.
Iwata T, Yamato M, Zhang Z, Mukobata S, Washio K, Ando T, Feijen J, Okano T, Ishikawa I. Validation of human periodontal ligament-derived cells as a reliable supply for cytotherapeutic use. J Clin Periodontol. 2010;37(12):1088-99. https://doi.org/10.1111/j.1600-051X.2010.01597.x.
Chen F-M, Gao L-N, Tian B-M, Zhang X-Y, Zhang Y-J, Dong G-Y, Lu H, et al. Treatment of periodontal intrabony defects using autologous periodontal ligament stem cells: a randomized clinical trial. Stem Cell Res Ther. 2016;7:33. https://doi.org/10.1186/s13287-016-0288-1.
Article PubMed PubMed Central Google Scholar
Bakopoulou A, Leyhausen G, Volk J, Tsiftsoglou A, Garefis P, Koidis P, Geurtsen W. Comparative evaluation of in vitro osteo/odontogenic differentiation potential of human dental pulp stem cells (DPSCs) and stem cells from the apical papilla (SCAP). Arch Oral Biol. 2011;56(7):709-21. https://doi.org/10.1016/j.archoralbio.2010.12.008.
Han C, Yang Z, Zhou W, Jin F, Song Y, Wang Y, Huo N, Chen L, Qian H, Hou R, Duan Y, Jin Y. Periapical follicle stem cell: a promising candidate for cementum/periodontal ligament regeneration and bio-root engineering. Stem Cells Dev. 2010;19(9):1405-15. https://doi.org/10.1089/scd.2009.0277.
Article CAS PubMed Google Scholar
Luan X, Ito Y, Dangaria S, Diekwisch TG. Dental follicle progenitor cell heterogeneity within the developing mouse periodontium. Stem Cells Dev. 2006;15(4):595-608. https://doi.org/10.1089/scd.2006.15.595.
Article CAS PubMed PubMed Central Google Scholar
Handa K, Saito M, Tsunoda A, Yamauchi M, Hattori S, Sato S, Toyoda M, Teranaka T, Narayanan AS. Progenitor cells from dental follicle are able to kind cementum matrix in vivo. Connect Tissue Res. 2002;43(2-3):406-eight PMID: 12489190.
Guo W, Chen L, Gong K, Ding B, Duan Y, Jin Y. Heterogeneous dental follicle cells and the regeneration of complicated periodontal tissues. Tissue Engineering. Part A. 2012;18(5-6):459-70 https://doi.org/10.1089/ten.tea.2011.0261.
Bai, Yudi et al. Cementum- and periodontal ligament-like tissue formation by dental follicle cell sheets co-cultured with Hertwig’s epithelial root sheath cells. Bone. 2011, 48, Issue 6, pp. 1417-1426, https://doi.org/10.1016/j.bone.2011.02.016.
Cordeiro MM, Dong Z, Kaneko T, Zhang Z, Miyazawa M, Shi S, et al. Dental pulp tissue engineering with stem cells from exfoliated deciduous teeth. 2008, 34, pp. 962-969.
Dobie K, Smith G, Sloan AJ, Smith AJ. Effects of alginate, hydrogels and TGF-beta 1 on human dental pulp repair in vitro. Connect Tissue Res 2. 2002;43:387-90.
Friedlander LT, Cullinan MP, Love RM. Dental stem cells and their potential position in apexogenesis and apexification. Int Endod J. 2009;42:955-62.
Cai J, Zhang Y, Liu P, Chen S, Wu X, Sun Y, Li A, Huang K, Luo R, Wang L, Liu Y, Zhou T, Wei S, Pan G, Pei D, Generation of tooth-like buildings from integration-free human urine induced pluripotent stem cells. Cell Regen (Lond). July 30, 2013, 2(1), pp. 6, doi: https://doi.org/10.1186/2045-9769-2-6.
Craig J. Taylor, Eleanor M. Bolton, and J. Andrew Bradley 2011 Aug 12 and https://doi.org/10.1098/rstb.2011.0030], 366(1575): 2312-2322. [doi:. Immunological concerns for embryonic and induced pluripotent stem cell banking,. Philos Trans R SocLond B Biol Sci. 2011, 366(1575), pp. 2312-2322, doi:https://doi.org/10.1098/rstb.2011.0030.
T.R. Nayak, H. Andersen, V.S. Makam, C. Khaw, S. Bae, X.F. Xu, P.L.R. Ee, J.H. Ahn, B.H. Hong, G. Pastorin, B. Ozyilmaz, ACS Nano, 5 (6) (2011), pp. 4. Graphene for managed and accelerated osteogenic differentiation of human mesenchymal stem cells,. ACS Nano. 2011, pp. 4670-4678.
Lee WC, Lim C, Shi H, Tang LAL, Wang Y, Lim CT, Loh KP. Origin of enhanced stem cell growth and differentiation on graphene and graphene oxide. ACS Nano. 2011;5(9):7334-41.
Kenry LWC, Loh KP, Lim CT. When stem cells meet graphene: alternatives and challenges in regenerative medication. Biomaterials. 2018;155:236-50.
Yuan A, Farber EL, Rapoport AL, Tejada D, Deniskin R, Akhmedov NB, et al. Transfer of microRNAs by embryonic stem cell microvesicles. 2009. 2009, 4(3), p. https://doi.org/10.1371/journal.pone. 0004722.
Oh, Myeongsik, et al. Exosomes derived from human induced pluripotent stem cells ameliorate the aging of skin fibroblasts. Int. J. Mol. Sci. 2018, 19(6), p. 1715.
Ramirez MI. et al. Technical challenges of working with extracellular vesicles. Nanoscale. 2018;10:881-906.
Valadi H, et al. Exosome-mediated switch of mRNAs and microRNAs is a novel mechanism of genetic trade between cells. Nat. Cell Biol. 2007;9:654-9.
Mateescu B, et al. Obstacles and opportunities within the purposeful evaluation of extracellular vesicle RNA-an ISEV position paper. J. Extracell. Vesicles. 2017;6(1). https://doi.org/10.1080/20013078.2017.1286095.
Nawaz M, et al. Extracellular vesicles: evolving components in stem cell biology. Stem Cells Int. 2016;2016:17. Article ID 1073140.
Helfrich, Y.R., Sachs, D.L. and Voorhees, J.J. Overview of skin aging and photoaging. Dermatol. Nurs. 20, pp. 177-183, https://www.ncbi.nlm.nih.gov/pubmed/18649702.
Julia Tigges, Jean Krutmann, Ellen Fritsche, Judith Haendeler, Heiner Schaal, Jens W. Fischer, Faiza Kalfalah, Hans Reinke, Guido Reifenberger, Kai Stühler, Natascia Ventura, Sabrina Gundermann, Petra Boukamp, Fritz Boege. The hallmarks of fibroblast ageing, mechanisms of ageing and growth, 138, 2014, Pages 26-44. 2014, 138, pp. 26-44, ISSN 0047-6374, https://doi.org/10.1016/j.mad.2014.03.004.
Huh MI, Kim MS, Kim HK, et al. Effect of conditioned media collected from human amniotic fluid-derived stem cells (hAFSCs) on pores and skin regeneration and photograph-aging. Tissue Eng Regen Med. 2014;11:171 https://doi.org/10.1007/s13770-014-0412-1.
Togel F, Hu Z, Weiss K, et al. Administered mesenchymal stem cells protect against ischemic acute renal failure by way of differentiation-impartial mechanisms. Am J Physiol Renal Physiol. 2005;289:F31.
Liu J, Han G, Liu H, et al. Suppression of cholangiocarcinoma cell growth by human umbilical cord mesenchymal stem cells: a potential function of Wnt and Akt signaling. PLoS One. 2013;8:e62844.
Oh M, et al. Promotive results of human induced pluripotent stem cell-conditioned medium on the proliferation and migration of dermal fibroblasts. Biotechnol. Bioprocess Eng. 2017;22:561-8.
Chen L, Tredget EE, Wu PY, Wu Y. Paracrine elements of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PloS One. 2008;3:e1886.
Bae J-S, Lee S-H, Kim J-E, Choi J-Y, Park R-W, Park JY, Park H-S, Sohn Y-S, Lee D-S, Lee EB. βig-h3 supports keratinocyte adhesion, migration, and proliferation by way of α3β1 integrin. Biochem. Biophys. Res. Commun. 2002;294:940-8.
Zhou B-R, Xu Y, Guo S-L, Xu Y, Wang Y, Zhu F, Permatasari F, Wu D, Yin Z-Q, Luo D. The effect of conditioned media of adipose-derived stem cells on wound healing after ablative fractional carbon dioxide laser resurfacing. BioMed Res. Int. 2013;519:126.
Peng Y, Baulier E, Ke Y, Young A, Ahmedli NB, Schwartz SD, et al. Human embryonic stem cells extracellular vesicles and their results on immortalized human retinal Müller cells. PLoS ONE. 2018, 13(3), p. https://doi.org/10.1371/journal.pone.019400.
Harris MT, Butler DL, Boivin GP, Florer JB, Schantz EJ, Wenstrup RJ. Mesenchymal stem cells used for rabbit tendon restore can form ectopic bone and specific alkaline phosphatase exercise in constructs. J Orthop Res. 2004;22:998-1003.
Mascetti VL, Pedersen RA. Human-mouse chimerism validates human stem cell pluripotency. Cell Stem Cell. 2016;18:67-72.
Gandia C, Armiñan A, García-Verdugo JM, Lledó E, Ruiz A, Miñana MD, Sanchez-Torrijos J, Payá R, Mirabet V, Carbonell-Uberos F, Llop M, Montero JA, Sepúlveda P. Human dental pulp stem cells improve left ventricular operate, induce angiogenesis, and cut back infarct dimension in rats with acute myocardial infarction. Stem Cells. 2007;26(3):638-45.
Perry BC, Zhou D, Wu X, Yang FC, Byers MA, Chu TM, Hockema JJ, Woods EJ, Goebel WS. Collection, cryopreservation, and characterization of human dental pulp-derived mesenchymal stem cells for banking and clinical use. Tissue Eng Part C Methods. 2008;14(2):149-56.
Garcia-Olmo D, Garcia-Arranz M, Herreros D, et al. A section I clinical trial of the therapy of Crohn’s fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48:1416-23.
de Mendonça CA, Bueno DF, Martins MT, Kerkis I, Kerkis A, Fanganiello RD, Cerruti H, Alonso N, Passos-Bueno MR. Reconstruction of massive cranial defects in nonimmunosuppressed experimental design with human dental pulp stem cells. J Craniofac Surg. 2008;19(1):204-10.
Google Scholar
Seo BM, Sonoyama W, Yamaza T, Coppe C, Kikuiri T, Akiyama K, Lee JS, Shi S. SHED restore vital-measurement calvarial defects in mice. Oral Dis. 2008;14(5):428-34.
Abbas, Diakonov I., Sharpe P. Neural crest origin of dental stem cells. Pan European Federation of the International Association for Dental Research (PEF IADR). 2008, Vols. Seq #96 – Oral Stem Cells.
Kerkis I, Ambrosio CE, Kerkis A, Martins DS, Gaiad TP, Morini AC, Vieira NM, Marina P, et al. Early transplantation of human immature dental pulp stem cells from baby teeth to golden retriever muscular dystrophy (GRMD) dogs. J Transl Med. 2008;6:35.
PubMed PubMed Central Google Scholar
Xianrui Yang, Li Li, Li Xiao, Donghui Zhang. Recycle the dental fairy’s bundle: overview of dental pulp stem cells. Stem Cell Res Ther. 2018, 9, 1, 1. https://doi.org/10.1186/s13287-018-1094-8.
Wang J, Wang X, Sun Z, Wang X, Yang H, Shi S, Wang S. Stem cells from human-exfoliated deciduous teeth can differentiate into dopaminergic neuron-like cells. Stem Cells Dev. 2010;19:1375-83.
Wang J, et al. The odontogenic differentiation of human dental pulp stem cells on nanofibrous poly (L-lactic acid) scaffolds in vitro and in vivo. Acta Biomater. 2010;6(10):3856-63.
Davies OG, Cooper PR, Shelton RM, Smith AJ, Scheven BA. A comparability of the in vitro mineralisation and dentinogenic potential of mesenchymal stem cells derived from adipose tissue, bone marrow and dental pulp. J Bone Miner Metab. 2015;33:371-82.
Huang GT-J, Shagramanova K, Chan SW. Formation of odontoblast-like cells from cultured human dental pulp cells on dentin in vitro. J Endod. 2006;32:1066-73.
Shi S, Robey PG, Gronthos S. Comparison of human dental pulp and bone marrow stromal stem cells by cDNA microarray evaluation. Bone. 2001;29(6):532-9.
Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U S A. 2000;97:13625-30.
Nuti N, Corallo C, Chan BMF, Ferrari M, Gerami-Naini B. Multipotent differentiation of human dental pulp stem cells: a literature evaluation. Stem Cell Rev Rep. 2016;12:511-23.
Ferro F, et al. Dental pulp stem cells differentiation reveals new insights in Oct4A dynamics. PloS One. 2012;7(7):e41774.
Conde MCM, Chisini LA, Grazioli G, Francia A, Carvalho RVd, Alcázar JCB, Tarquinio SBC, Demarco FF. Does cryopreservation have an effect on the biological properties of stem cells from dental tissues? A systematic evaluate. Braz Dent J. 2016;1210(6):633-40. https://doi.org/10.1590/0103-6440201600980.
Papaccio G, Graziano A, d’Aquino R, Graziano MF, Pirozzi G, Menditti D, De Rosa A, Carinci F, Laino G. Long-term cryopreservation of dental pulp stem cells (SBP-DPSCs) and their differentiated osteoblasts: a cell supply for tissue repair. J Cell Physiol. 2006;208:319-25.
Alge DL, Zhou D, Adams LL, et. al. Donor-matched comparability of dental pulp stem cells and bone marrow-derived mesenchymal stem cells in a rat model. J Tissue Eng Regen Med. 2010;4(1):73-81.
Jo Y-Y, Lee H-J, Kook S-Y, Choung H-W, Park J-Y, Chung J-H, Choung Y-H, Kim E-S, Yang H-C, Choung P-H. Isolation and characterization of postnatal stem cells from human dental tissues. Tissue Eng. 2007;13:767-73.
Gronthos S, Brahim J, Li W, Fisher LW, Cherman N, Boyde A, DenBesten P, Robey PG, Shi S. Stem cell properties of human dental pulp stem cells. J Dent Res. 2002;81:531-5.
Laino G, d’Aquino R, Graziano A, Lanza V, Carinci F, Naro F, Pirozzi G, Papaccio G. A new population of human adult dental pulp stem cells: a useful source of dwelling autologous fibrous bone tissue (LAB). J Bone Miner Res. 2005;20:1394-402.
Zainal A, Shahrul H, et al. In vitro chondrogenesis transformation examine of mouse dental pulp stem cells. Sci World J. 2012;2012:827149.
Wei X, et al. Expression of mineralization markers in dental pulp cells. J Endod. 2007;33(6):703-8.
Dai J, et al. The effect of co-culturing costal chondrocytes and dental pulp stem cells mixed with exogenous FGF9 protein on chondrogenesis and ossification in engineered cartilage. Biomaterials. 2012;33(31):7699-711.
Vasandan AB, et al. Functional variations in mesenchymal stromal cells from human dental pulp and periodontal ligament. J Cell Mol Med. 2014;18(2):344-54.
CAS PubMed PubMed Central Google Scholar
Werle SB, et al. Carious deciduous teeth are a possible source for dental pulp stem cells. Clin Oral Investig. 2015;20:75-81.
Nemeth CL, et al. Enhanced chondrogenic differentiation of dental pulp stem cells using nanopatterned PEG-GelMA-HA hydrogels. Tissue Eng A. 2014;20(21-22):2817-29.
CAS Google Scholar
Paino F, Ricci G, De Rosa A, D’Aquino R, Laino L, Pirozzi G, et al. Ecto-mesenchymal stem cells from dental pulp are dedicated to differentiate into active melanocytes. Eur. Cell Mater. 2010;20:295-305.
CAS PubMed Google Scholar
Ferro F, Spelat R, Baheney CS. Dental pulp stem cell (DPSC) isolation, characterization, and differentiation. In: Kioussi C, editor. Stem cells and tissue restore. Methods in molecular biology (methods and protocols): Humana Press. 2014;1210.
Ishkitiev N, Yaegaki K, Imai T, Tanaka T, Nakahara T, Ishikawa H, Mitev V, Haapasalo M. High-purity hepatic lineage differentiated from dental pulp stem cells in serum-free medium. J Endod. 2012;38:475-80.
PubMed Google Scholar
Download references
Acknowledgements
Funding
This work is supported by Wrocław Medical University in Poland.
Availability of knowledge and materials
Please contact creator for knowledge requests.
Author info
Authors and Affiliations
Department of Experimental Surgery and Biomaterials Research, Wroclaw Medical University, Bujwida 44, Wrocław, 50-345, Poland
Wojciech Zakrzewski, Maria Szymonowicz & Zbigniew Rybak
Department of Conservative Dentistry and Pedodontics, Krakowska 26, Wrocław, 50-425, Poland
Maciej Dobrzyński
Wojciech ZakrzewskiView author publicationsYou can too search for this writer in PubMed Google Scholar
Maciej DobrzyńskiView author publicationsYou may also search for this writer in PubMed Google Scholar
Maria SzymonowiczView creator publicationsYou can also seek for this creator in PubMed Google Scholar
Zbigniew RybakView author publicationsYou may as well seek for this writer in PubMed Google Scholar
Contributions
WZ is the principal author and was responsible for the primary draft of the manuscript. WZ and ZR were accountable for the idea of the assessment. MS, MD, and ZR had been chargeable for revising the article and for knowledge acquisition. All authors read and accepted the ultimate manuscript.
Corresponding creator
Correspondence to Wojciech Zakrzewski.
Ethics declarations
Ethics approval and consent to participate
Consent for publication
Not applicable.
Competing interests
The authors declare that they don’t have any competing interests.
Publisher’s Note
Springer Nature stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This text is distributed beneath the terms of the Creative Commons Attribution 4.Zero International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, offered you give acceptable credit score to the unique writer(s) and the supply, provide a link to the Creative Commons license, and indicate if changes have been made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the information made accessible in this article, except otherwise said.
Reprints and permissions
About this text
Cite this text
Zakrzewski, W., Dobrzyński, M., Szymonowicz, M. et al. Stem cells: past, present, and future. Stem Cell Res Ther 10, sixty eight (2019).