Category Archives: Induced Pluripotent Stem Cells

Clinical potential of human-induced pluripotent stem cells …

The recent establishment of induced pluripotent stem (iPS) cells promises the development of autologous cell therapies for degenerative diseases, without the ethical concerns associated with human embryonic stem (ES) cells. Initially, iPS cells were generated by retroviral transduction of somatic cells with core reprogramming genes. To avoid potential genotoxic effects associated with retroviral transfection, more recently, alternative non-viral gene transfer approaches were developed. Before a potential clinical application of iPS cell-derived therapies can be planned, it must be ensured that the reprogramming to pluripotency is not associated with genome mutagenesis or epigenetic aberrations. This may include direct effects of the reprogramming method or "off-target" effects associated with the reprogramming or the culture conditions. Thus, a rigorous safety testing of iPS or iPS-derived cells is imperative, including long-term studies in model animals. This will include not only rodents but also larger mammalian model species to allow for assessing long-term stability of the transplanted cells, functional integration into the host tissue, and freedom from undifferentiated iPS cells. Determination of the necessary cell dose is also critical; it is assumed that a minimum of 1 billion transplantable cells is required to achieve a therapeutic effect. This will request medium to long-term in vitro cultivation and dozens of cell divisions, bearing the risk of accumulating replication errors. Here, we review the clinical potential of human iPS cells and evaluate which are the most suitable approaches to overcome or minimize risks associated with the application of iPS cell-derived cell therapies.

Read more from the original source:
Clinical potential of human-induced pluripotent stem cells ...

Why Induced Pluripotent Stem Cells Are Vital for Glaucoma …

One of the most significant discoveries in regeneration research occurred when scientists learned that mature stem cells could be reprogrammedturned back into young stem cellsthen used to grow any type of new tissue. This revelation changed everything the experts thought they knew about cell development. Until then, they hadnt dreamed they could turn back the hands of time in old stem cells.

Now stem cells are poised to make a similar impact on glaucoma research and treatment, as adult stem cells can be taken from the eye or skin, and used to try to replace damaged cells in your eye. Stem cell treatment may one day restore vision lost to glaucoma, so this is a topic glaucoma patients and research supporters will want to know about.

At the start of life, embryonic stem cells are pluripotent, which means they have the remarkable ability to become any type of cell in the body. Following conception, stem cells rapidly reproduce to form clusters of cells that begin to specialize, or differentiate. Each cluster follows a different path, developing into the heart, brain, lungs, skin and every other tissue needed to build the human body.

Fully mature, adult stem cells continue to generate new cells, but only for the specific tissues where they live. For example, hair follicles have adult stem cells that regrow hair and adult stem cells in bone marrow give rise to blood cells, but they cant fill in for one another. These mature stem cells are the sentinels that guard your health, as they replace cells that are damaged due to normal wear-and-tear, injury and disease.

As long as theyre alive and thriving, adult stem cells continue to self-renew indefinitely, dividing and replicating as often as needed. Even if theyre inactive for a long time, they can jump back into action at a moments notice. But theres one thing they cant do: they cant reverse back into their pluripotent state. At least, they cant do that in their natural environment.

In the early 1960s, Sir John Bertrand Gurdon was a young developmental biologist searching for the answer to one question: Is it possible for adult stem cells to return to their immature state? He experimented with frog cells, transplanting mature stem cells into eggs that had their stem cells removed. After many trials, an astonishing thing happenedthe eggs grew into normal tadpoles. With that success, Gurdon proved that fully-differentiated adult stem cells retained the genetic information found in pluripotent embryonic cells.1

Nearly 50 years later, Shinya Yamanaka, MD, PhD and his co-workers published a stunning study. In a long series of experiments, he isolated 24 genes responsible for pluripotency. Then he reintroduced these genes into mature stem cells, individually and in various combinations, until he narrowed it down to four key genes. When used together, the four genes, now dubbed Yamanaka factors, accomplished the unbelievablethey reprogrammed adult stem cells, making them convert back into embryonic stem cells. The induced pluripotent stem cell had been discovered.2

Gurdon and Yamanaka were jointly awarded the 2012 Nobel Prize in Physiology or Medicine for these two discoveries.3 Of course, they both continued to study stem cells and, combined with results from other experts in the field, significant progress has been made. Now induced pluripotent stem cellsor iPS cells for shortcan be formed from human cells and they have a leading role in glaucoma research. Glaucoma Research Foundation gave our 2015 Visionary Award to Dr. Yamanaka to honor his pioneering work to improve global healthcare and treat blinding eye disease.

You may begin to hear a lot about iPS cells being used to develop treatments for glaucoma. When damaged cells in an area called the trabecular meshwork are replaced with iPS cells, intraocular pressure is normalized. If iPS cells could be used to restore parts of the retina, like photoreceptor, ganglion and Muller cells, vision could be restored. Heres one version of how the process might look:

A doctor takes a sample of cells called fibroblasts from a small area of skin on your arm. The fibroblasts are sent to a lab, put into a glass petri dish and injected with Yamanaka factors that convert them into induced pluripotent stem cells. Then substances known to trigger differentiation are added to the cells. They may be directed to become retinal ganglion cells, trabecular meshwork cells or another targeted cell in the eye. When a sufficient number of specialized cells are ready, theyre injected into the damaged eye, where they continue to grow and facilitate healing.4

This scenario isnt entirely hypothetical. Research using iPS cells to treat glaucoma is still in the early stages, but the European Commission has already authorized stem cell treatment for injured corneas. Their decision was based on clinical trials showing that healthy limbal stem cells could be taken from the cornea, expanded in the lab and transplanted back into the damaged part of the eye. The new iPS cells safely and effectively repaired the cornea and restored vision.5

Researchers also use iPS cells to create models of human cells and use them to learn how glaucoma progresses and to test emerging pharmaceutical treatments. Some of the most promising research uses iPS cells to develop models of retinal ganglion cells.6 Ganglion cells collect visual input and send it to the brainmore than a million ganglion cells are bundled together to form the optic nerveand theyre preferentially vulnerable among all retinal cells to glaucoma-inflicted damage.

Beyond their versatile uses, iPS cells have two other critical benefits. They allow glaucoma researchers to pursue new treatments while avoiding ethical concerns related to using embryonic stem cells. And best of all, when mature stem cells come from the same person who will use them for treatment, they dont have to worry about rejection by the immune system because the cells are already a genetic match. This is personalized medicine at a whole new level.

Much research has yet to be done, and clinical trials to test stem cell procedures on people with glaucoma are still down the road, yet the work accomplished so far shines enough light to show that answers are within reach. Glaucoma Research Foundation is determined to support research that will one day make the promise of restored vision come true.

Glaucoma Research Foundation depends on your donation to support research and patient education. Learn about the many ways you can join our cause.

More:
Why Induced Pluripotent Stem Cells Are Vital for Glaucoma ...

Live Cell Imaging of Induced Pluripotent Stem Cell …

Our live cell image program supports the advancement of iPSC technology in three ways:

1) Identification of process control measurements: A critical component to the translation of iPSCs into therapeutic applications is to design principles for predictably and reproducibly culturing cells and efficiently differentiating them into cell types of interest. Live cell imaging provides high-resolution measurements in the sense that we collect time-dependent data from large numbers of individual cells. We then use this data to discover lower resolution measurements, such as the activity of a biomarker at a single point in time, that can serve as critical process control points during processing of pluripotent stem cells.

2) Interpreting biomarkers: Cells are stochastic and dynamic and may interconvert between states and the expression of biomarkers can change over time. The predictive power of a biomarker or a set of biomarkers the indicate the differentiated state of a cell can be evaluated by examining the history of that cell by tracking forward and backward in time through a time lapse image set.

3) Predictive modeling: We have shown that fluctuations in promoter activity can be used in combination with appropriate models to predict rates of state change in cell populations. Similar mathematical models that can inform bioprocessing decisions during scale-up will be critical to obtaining iPSC populations with a desired set of characteristics.

Over the past several years, we have developed tools for measuring parameters related to size, shape and intensity from single cells over time (Halter Cytometry 2011). We have also developed modeling tools for using the temporal information to model the stochastic and deterministic components of gene expression (Sisan PNAS 2012; Lund Phys Chem B 2014).

We are now applying these live cell imaging tools to the study of stem cell pluripotency and differentiation (Bhadriraju Stem Cell Research 2016). Induced pluripotent stem cell technologies are a powerful new tool for biomedical research and have the potential to revolutionize medicine.

Excerpt from:
Live Cell Imaging of Induced Pluripotent Stem Cell ...

Induced Pluripotent Stem Cells – cellapplications.com

Documented Consistency

Quality Control and Testing

Product Selection & Support

HiPSC Custom Services

For Research Use Only - Not for Human or Clinical Applications

Human Induced Pluripotent Stem Cells (HiPSC) Top: HiPSC express pluriotency markers OCT4, Nanog, LIN28 and SSEA-4. Bottom: HiPSC differentiate into cell derivatives from the 3 embryonic layers: Neuronal marker beta III tubulin (TUJ1), Smooth Muscle Actin (SMA) and Hepatocyte Nuclear Factor 3 Beta (HNF3b).

Cell Applications, Inc. has a deep, rich history in HiPSC

Human Dermal Fibroblasts (HDF) from Cell Applications were used by Nobel Laureate S. Yamanaka to establish iPSC in his groundbreaking publications in 2007, unleashing a revolution in stem cell biology. Yamanaka and collaborators demonstrated that expression of four transcription factors widely prevalent in embryonic stem cells is sufficient to trigger the transition of somatic cells towards a pluripotent state that resembles embryonic stem cells in many aspects, such as the expression of classic pluripotency markers and the ability to generate cell derivatives from the three embryonic germ layers.

HiPSC are generated from somatic cells, eliminating ethical considerations associated with scientific work based on embryonic stem cells. Furthermore, being donor/patient-specific, they open possibilities for a wide variety of studies in biomedical research. Donor somatic cells carry the genetic makeup of the diseased patient, hence HiPSC can be used directly to model disease on a dish.

Thus, one of the main uses of HiPSC has been in genetic disease modeling in organs and tissues, such as the brain (Alzheimers, Autism Spectrum Disorders), heart (Familial Hypertrophic, Dilated, and Arrhythmogenic Right Ventricular Cardiomyopathies), and skeletal muscle (Amyotrophic Lateral Sclerosis, Spinal Muscle Atrophy). The combination of HiPSC technology and gene editing strategies such as the CRISPR/Cas9 system creates a powerful platform in which disease-causing mutations can be created on demand and sets of isogenic cell lines (with and without mutations) serve as convenient tools for disease modeling studies.

Other applications of HiPSC and iPSC-differentiated cells include drug screening, development, efficacy and toxicity assessment. As an example, through the FDA-backed CiPA (Comprehensive in vitro Pro-Arrhythmia Assessment) initiative, HiPSC-derived cardiac muscle cells (cardiomyocytes) are poised to constitute a new standard model for the evaluation of cardiotoxicity of new drugs, which is the main reason of drug withdrawal from the market. Finally, HiPSC-differentiated cells are being used in early stage technology development for applications in regenerative medicine. Bio-printing and tissue constructs have also been considered as attractive applications for HiPSC.

StemoniX

Our partner StemoniX is a cutting-edge biotechnology company that is leading the development and manufacturing of HiPSC. They generate biologically accurate miniaturized organ microtissue for academic and industrial pharmaceutical research and discovery. StemoniX, a licensee of Academia Japans iPS patent portfolio, provides high quality HiPSC cells to researchers around the world. StemoniX HiPSC are thoroughly characterized for pluripotency with established pluripotency markers. Proven technology incorporating the latest innovations is able to provide cardiomyocytes with in vitro-like features. Confirmative tests show the HiPSC differentiate into derivatives from the 3 embryonic layers.

Press & Media Relations

Daniel Schroen, PhD

VP, Sales & Marketing

(858) 453-0848

Here is the original post:
Induced Pluripotent Stem Cells - cellapplications.com

Induced pluripotent stem cell models from X-linked …

Objective:

Because of a lack of an appropriate animal model system and the inaccessibility of human oligodendrocytes in vivo, X-linked adrenoleukodystrophy (X-ALD)-induced pluripotent stem cells (iPSCs) would provide a unique cellular model for studying etiopathophysiology and development of therapeutics for X-ALD.

We generated and characterized iPSCs of the 2 major types of X-ALD, childhood cerebral ALD (CCALD) and adrenomyeloneuropathy (AMN), and differentiated them into oligodendrocytes and neurons. We evaluated disease-relevant phenotypes by pharmacological and genetic approaches.

We established iPSCs from the patients with CCALD and AMN. Both CCALD and AMN iPSCs normally differentiated into oligodendrocytes, the cell type primarily affected in the X-ALD brain, indicating no developmental defect due to the ABCD1 mutations. Although low in X-ALD iPSCs, very long chain fatty acid (VLCFA) level was significantly increased after oligodendrocyte differentiation. VLCFA accumulation was much higher in CCALD oligodendrocytes than AMN oligodendrocytes but was not significantly different between CCALD and AMN neurons, indicating that the severe clinical manifestations in CCALD might be associated with abnormal VLCFA accumulation in oligodendrocytes. Furthermore, the abnormal accumulation of VLCFA in the X-ALD oligodendrocytes can be reduced by the upregulated ABCD2 gene expression after treatment with lovastatin or 4-phenylbutyrate.

X-ALD iPSC model recapitulates the key events of disease development (ie, VLCFA accumulation in oligodendrocytes), provides new clues for better understanding of the disease, and allows for early and accurate diagnosis of the disease subtypes. X-ALD oligodendrocytes can be a useful cell model system to develop new therapeutics for treating X-ALD. ANN NEUROL 2011;

Originally posted here:
Induced pluripotent stem cell models from X-linked ...

Generation of germline-competent induced pluripotent stem …

We have previously shown that pluripotent stem cells can be induced from mouse fibroblasts by retroviral introduction of Oct3/4 (also called Pou5f1), Sox2, c-Myc and Klf4, and subsequent selection for Fbx15 (also called Fbxo15) expression. These induced pluripotent stem (iPS) cells (hereafter called Fbx15 iPS cells) are similar to embryonic stem (ES) cells in morphology, proliferation and teratoma formation; however, they are different with regards to gene expression and DNA methylation patterns, and fail to produce adult chimaeras. Here we show that selection for Nanog expression results in germline-competent iPS cells with increased ES-cell-like gene expression and DNA methylation patterns compared with Fbx15 iPS cells. The four transgenes (Oct3/4, Sox2, c-myc and Klf4) were strongly silenced in Nanog iPS cells. We obtained adult chimaeras from seven Nanog iPS cell clones, with one clone being transmitted through the germ line to the next generation. Approximately 20% of the offspring developed tumours attributable to reactivation of the c-myc transgene. Thus, iPS cells competent for germline chimaeras can be obtained from fibroblasts, but retroviral introduction of c-Myc should be avoided for clinical application.

Here is the original post:
Generation of germline-competent induced pluripotent stem ...

Induced pluripotent stem-cell therapy – Wikipedia

In 2006, Shinya Yamanaka of Kyoto University in Japan was the first to disprove the previous notion that reversible cell differentiation of mammals was impossible. He reprogrammed a fully differentiated mouse cell into a pluripotent stem cell by introducing four genes, Oct-4, SOX2, KLF4, and Myc, into the mouse fibroblast through gene-carrying viruses. With this method, he and his coworkers created induced pluripotent stem cells (iPS cells), the key component in this experiment.[1] Scientists have been able to conduct experiments that show the ability of iPS cells to treat and even cure diseases. In this experiment, tests were run on mice with inherited sickle-cell anemia. Skin cells were turned into cells containing genes that transformed the cells into iPS cells. These replaced the diseased sickled cells, curing the test mice. The reprogramming of the pluripotent stem cells in mice was successfully duplicated with human pluripotent stem cells within about a year of the experiment on the mice.[citation needed]

Sickle-cell anemia is a disease in which the body produces abnormally shaped red blood cells. Red blood cells are flexible and round, moving easily through the blood vessels. Infected cells are shaped like a crescent or sickle (the namesake of the disease). As a result of this disorder the hemoglobin protein in red blood cells is faulty. Normal hemoglobin bonds to oxygen, then releases it into cells that need it. The blood cell retains its original form and is cycled back to the lungs and re-oxygenated.

Sickle cell hemoglobin, however, after giving up oxygen, cling together and make the red blood cell stiff. The sickle shape also makes it difficult for the red blood cell to navigate arteries and causes blockages.[2] This can cause intense pain and organ damage. The sickled red blood cells are fragile and prone to rupture. When the number of red blood cells decreases from rupture (hemolysis), anemia is the result. Sickle cells die in 1020 days as opposed to the traditional 120-day lifespan of a normal red blood cell.

Sickle cell anemia is inherited as an autosomal (meaning that the gene is not linked to a sex chromosome) recessive condition.[2] This means that the gene can be passed on from a carrier to his or her children. In order for sickle cell anemia to affect a person, the gene must be inherited from both the mother and the father, so that the child has two recessive sickle cell genes (a homozygous inheritance). People who inherit one sickle cell gene from one parent and one normal gene from the other parent, i.e. heterozygous patients, have a condition called sickle cell trait. Their bodies make both sickle hemoglobin and normal hemoglobin. They may pass the trait on to their children.

The effects of sickle-cell anemia vary from person to person. People who have the disease suffer from varying degrees of chronic pain and fatigue. With proper care and treatment, the quality of health of most patients will improve. Doctors have learned a great deal about sickle cell anemia since its discovery in 1979. They know its causes, its effects on the body, and possible treatments for complications. Sickle cell anemia has no widely available cure. A bone marrow transplant is the only treatment method currently recognized to be able to cure the disease, though it does not work for every patient. Finding a donor is difficult and the procedure could potentially do more harm than good. Treatments for sickle cell anemia are generally aimed at avoiding crises, relieving symptoms, and preventing complications. Such treatments may include medications, blood transfusions, and supplemental oxygen.

During the first step of the experiment, skin cells (also known as fibroblasts) were collected from infected test mice and put in a culture. The fibroblasts were reprogrammed by infecting them with retroviruses that contained genes common to embryonic stem cells. These genes were the same four used by Yamanaka (Oct-4, SOX2, KLF4, and Myc) in his earlier study. The investigators were trying to produce cells with the potential to differentiate into any type of cell needed (i.e. pluripotent stem cells). As the experiment continued, the fibroblasts multiplied into identical copies of iPS cells. The cells were then treated to form the mutation needed to reverse the anemia in the mice. This was accomplished by restructuring the DNA containing the defective globin gene into DNA with the normal gene through the process of homologous recombination. The iPS cells then differentiated into blood stem cells, or hematopoietic stem cells. The hematopoietic cells were injected back into the infected mice, where they proliferate and differentiate into normal blood cells, curing the mice of the disease.[3][4][verification needed]

To determine whether the mice were cured from the disease, the scientists checked for the usual symptoms of sickle cell disease. They examined the blood for mean corpuscular volume (MCV) and red cell distribution width (RDW) and urine concentration defects. They also checked for sickled red blood cells. They examined the DNA through gel electrophoresis, checking for bands that display an allele that causes sickling. Compared to the untreated mice with the disease, which they used as a control, "the treated animals had marked increases in RBC counts, healthy hemoglobin, and packed cell volume levels".[5]

Researchers examined "the urine concentration defect, which results from RBC sickling in renal tubules and consequent reduction in renal medullary blood flow, and the general deteriorated systemic condition reflected by lower body weight and increased breathing."[5] They were able to see that these parts of the body of the mice had healed or improved. This indicated that "all hematological and systemic parameters of sickle cell anemia improved substantially and were comparable to those in control mice."[5] They cannot say if this will work in humans because a safe way to inject the genes for the induced pluripotent cells is still needed.[citation needed]

The reprogramming of the induced pluripotent stem cells in mice was successfully duplicated in humans within a year of the successful experiment on the mice. This reprogramming was done in several labs and it was shown that the iPS cells in humans were almost identical to original embryonic stem cells (ES cells) that are responsible for the creation of all structures in a fetus.[1] An important feature of iPS cells is that they can be generated with cells taken from an adult, which would circumvent many of the ethical problems associated with working with ES cells. These iPS cells also have potential in creating and examining new disease models and developing more efficient drug treatments.[6] Another feature of these cells is that they provide researchers with a human cell sample, as opposed to simply using an animal with similar DNA, for drug testing.

One major problem with iPS cells is the way in which the cells are reprogrammed. Using gene-carrying viruses has the potential to cause iPS cells to develop into cancerous cells.[1] Also, an implant made using undifferentiated iPS cells, could cause a teratoma to form. Any implant that is generated from using these iPS cells would only be viable for transplant into the original subject that the cells were taken from. In order for these iPS cells to become viable in therapeutic use, there are still many steps that must be taken.[5][7]

In the future, researchers hope that induced pluripotent cells may be used to treat other diseases. Pluripotency is a crucial part of disease treatment because iPS cells are capable of differentiation in a way that is very similar to embryonic stem cells which can grow into fully differentiated tissues. iPS cells also demonstrate high telomerase activity and express human telomerase reverse transcriptase, a necessary component in the telomerase protein complex. Also, iPS cells expressed cell surface antigenic markers expressed on ES cells. Also, doubling time and mitotic activity are cornerstones of ES cells, as stem cells must self-renew as part of their definition. As said, iPS cells are morphologically similar to embryonic stem cells. Each cell has a round shape, a large nucleolus and a small amount of cytoplasm. One day, the process may be used in practical settings to provide a fundamental way of regeneration.

See original here:
Induced pluripotent stem-cell therapy - Wikipedia

Generation of Neural Crest-Like Cells From Human …

Neural crest cells (NCC) hold great promise for tissue engineering, however the inability to easily obtain large numbers of NCC is a major factor limiting their use in studies of regenerative medicine. Induced pluripotent stem cells (iPSC) are emerging as a novel candidate that could provide an unlimited source of NCC. In the present study, we examined the potential of neural crest tissue-derived periodontal ligament (PDL) iPSC to differentiate into neural crest-like cells (NCLC) relative to iPSC generated from a non-neural crest derived tissue, foreskin fibroblasts (FF). We detected high HNK1 expression during the differentiation of PDL and FF iPSC into NCLC as a marker for enriching for a population of cells with NCC characteristics. We isolated PDL iPSC- and FF iPSC-derived NCLC, which highly expressed HNK1. A high proportion of the HNK1-positive cell populations generated, expressed the MSC markers, whilst very few cells expressed the pluripotency markers or the hematopoietic markers. The PDL and FF HNK1-positive populations gave rise to smooth muscle, neural, glial, osteoblastic and adipocytic like cells and exhibited higher expression of smooth muscle, neural, and glial cell-associated markers than the PDL and FF HNK1-negative populations. Interestingly, the HNK1-positive cells derived from the PDL-iPSC exhibited a greater ability to differentiate into smooth muscle, neural, glial cells and adipocytes, than the HNK1-positive cells derived from the FF-iPSC. Our work suggests that HNK1-enriched NCLC from neural crest tissue-derived iPSC more closely resemble the phenotypic and functional hallmarks of NCC compared to the HNK1-low population and non-neural crest iPSC-derived NCLC. J. Cell. Physiol. 232: 402-416, 2017. 2016 Wiley Periodicals, Inc.

2016 Wiley Periodicals, Inc.

See the original post here:
Generation of Neural Crest-Like Cells From Human ...

Generation of Induced Pluripotent Stem Cells with …

Induced pluripotent stem cells (iPSCs) share many characteristics with embryonic stem cells, but lack ethical controversy. They provide vast opportunities for disease modeling, pathogenesis understanding, therapeutic drug development, toxicology, organ synthesis, and treatment of degenerative disease. However, this procedure also has many potential challenges, including a slow generation time, low efficiency, partially reprogrammed colonies, as well as somatic coding mutations in the genome. Pioneered by Shinya Yamanaka's team in 2006, iPSCs were first generated by introducing four transcription factors: Oct 4, Sox 2, Klf 4, and c-Myc (OSKM). Of those factors, Klf 4 and c-Myc are oncogenes, which are potentially a tumor risk. Therefore, to avoid problems such as tumorigenesis and low throughput, one of the key strategies has been to use other methods, including members of the same subgroup of transcription factors, activators or inhibitors of signaling pathways, microRNAs, epigenetic modifiers, or even differentiation-associated factors, to functionally replace the reprogramming transcription factors. In this study, we will mainly focus on the advances in the generation of iPSCs with substitutes for OSKM. The identification and combination of novel proteins or chemicals, particularly small molecules, to induce pluripotency will provide useful tools to discover the molecular mechanisms governing reprogramming and ultimately lead to the development of new iPSC-based therapeutics for future clinical applications.

Original post:
Generation of Induced Pluripotent Stem Cells with ...

Induced pluripotent stem cells and Parkinson’s disease …

Review Authors

Correspondence: T. Wang, Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Road, Wuhan 430022, Hubei, China; E-mail: wangtaowh@hust.edu.cn

Many neurodegenerative disorders, such as Parkinson's disease (PD), are characterized by progressive neuronal loss in different regions of the central nervous system, contributing to brain dysfunction in the relevant patients. Stem cell therapy holds great promise for PD patients, including with foetal ventral mesencephalic cells, human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Moreover, stem cells can be used to model neurodegenerative diseases in order to screen potential medication and explore their mechanisms of disease. However, related ethical issues, immunological rejection and lack of canonical grafting protocols limit common clinical use of stem cells. iPSCs, derived from reprogrammed somatic cells, provide new hope for cell replacement therapy. In this review, recent development in stem cell treatment for PD, using hiPSCs, as well as the potential value of hiPSCs in modelling for PD, have been summarized for application of iPSCs technology to clinical translation for PD treatment.

adenoviral iPSCs

cluster of differentiation

cynomolgus macaque

dopamine

dopamine transporter

deep brain stimulation

flow cytometric analysis

glucocerebrosidase

human embryonic stem cells

human induced pluripotent stem cells

Lewy bodies

leucine-rich repeat kinase 2

mouse embryonic fibroblasts

major histocompatibility complex

mitochondrial DNA

neural stem cells

Parkinson's disease

subgranular zone

-synuclein

substantia nigra pars compacta

subventricular zone

valproic acid

zinc finger nuclease

zonisamide

Parkinson's disease (PD) is the second most common neurodegenerative disorder, and concerns progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) of the midbrain [1]. The crucial pathological feature of PD is presence of Lewy bodies (LBs), which are abnormal aggregates of -synuclein (SNCA) protein. Reported standardized incidence rates of PD are 818 per 100 000 person-years worldwide [2]. In China, prevalence of PD for those aged 65 years is 1.7% [3]. PD patients suffer from motor symptoms such as rest tremor, bradykinesia, rigidity and abnormal gait. Non-motor symptoms, such as olfactory dysfunction, psychiatric changes and sleep disorders, further impair PD patients' quality of life. Up to now, multiple factors have been found to involve pathogenesis of PD, including genetic susceptibility, environmental toxins, interruption of autophagy, neuroinflammation and most importantly advancing age. Although the precise mechanisms underlying the pathogenesis of PD are not well understood, interactions within these pathogenic factors give rise to loss of DA neurons within the SNpc. Unfortunately, current pharmacological and surgical treatments provide only insufficient symptomatic relief, but cannot reverse nor slow down the underlying loss of midbrain DA neurons. Stem cell transplantation, however, holds great promise in the treatment of PD.

Neural stem cells (NSCs) provide a potential endogenous source for neuron replacement therapy in neurodegenerative disorders such as PD. One of the most important essential features of NSCs is their proliferation potential. It has already been indicated that NSCs can differentiate directly into DA neurons and Suksuphew and Noisa have shown that they have high possibility for producing two undifferentiated daughter cells at early stages of development (symmetric division), and later cell division for production of differentiated neurons plus glial cells (asymmetric division) [4]. A further feature of NSCs' multipotency is their potential to differentiate into astrocytes and oligodendrocytes, as well as into neurons [5, 6]. NSCs in the subventricular zone (SVZ) can differentiate into olfactory neurons, while those of the subgranular zone (SGZ) can differentiate into granular neurons of the dentate gyrus [7]. Furthermore, when implanted into developing eyes, hippocampal NSCs have exhibited several morphological and immunological properties of retinal cells, including photoreceptors [7]. Differentiation of adult NSCs can be influenced by their local environment as well as by intrinsic programmes [8].

Human ESCs were the first human stem cells to be identified and cultured, by Thomson et al. in 1998 [9], and were proven at that time to be self-renewing and pluripotent. These properties indicated hESCs as having great promise for cell transplantation therapy. However, ethical concerns arose immediately as generation of hESCs requires destruction of the fertilized human embryo. A further significant problem with transplanting stem cells is associated with immunological rejection after transplantation of specific cells derived from allogeneic hESCs. In 2006, Yamanaka et al. reported generation of ES-like pluripotent stem cells from somatic fibroblasts, the so-called iPSCs [10]. Since then, many methods have been explored to generate hiPSCs from a wide variety of easily accessible source tissues, including skin, adipose and blood cells [11-14]. Unlike hESCs, there are no ethical issues preventing use of iPSCs. Refinement of reprogramming methods now allows for iPSC generation without genomic integration of reprogramming factors, using expression plasmids, non-integrating viruses, recombinant proteins, small molecules and synthetically modified mRNAs or miRNAs [15]. Here, we review the existing iPSC-based models and treatments, with particular emphasis on PD, and explore the challenges associated with cell therapies using iPSCs-derived DA neurons, which have thus far hindered expansion of this research.

Cell replacement therapy with foetal ventral midbrain (VM) DA neurons has been shown, in some ways, to be beneficial to PD patients. Dopaminergic neurons lost in PD are primarily of the VM, and VM DA neurons arise from floor plate cells during embryonic development. The earliest study of DA neuron differentiation from mouse ESCs was performed by Lee et al. in 2000 [16]. This group generated CNS progenitor populations from ESCs, expanded the cells and promoted their differentiation into dopaminergic and serotonergic neurons in the presence of mitogens and specific signalling molecules. Their differentiation involved a number of steps: generation of embryoid bodies (EBs) without retinoic acid (RA) treatment in a serum-containing medium, use of a defined medium to select for CNS stem cells, proliferation of CNS stem cells in the presence of mitogen, basic fibroblast growth factor (bFGF), and differentiation of the stem cells by removal of the mitogen in serum-free medium [16]. Finally, the differentiation medium consisted of N2 medium supplemented with laminin, cAMP and ascorbic acid (AA). Sonic hedgehog (SHH), FGF8 and AA also enhanced differentiation to DA fate, and increased yield of ES-derived TH+ neurons. The cells were incubated under differentiation conditions for 615 days at the last stage, in order to increase numbers of TH+ neurons and DA level [16]. While this protocol succeeded in generating DA neurons at relatively high efficiency, extensive studies in Parkinsonian animals are needed to further assess complete function and safety of ESC-derived DA neurons in vivo [16]. Efficiency and purification of generated cell populations also needs to be improved by genetic methods.

After some months, Kawasaki and colleagues introduced an efficient method for generating neurons from ESCs by using PA6-derived stromal cell-derived inducing activity (SDIA) in a serum-free condition requiring neither EBs nor RA treatment [17]. High proportions of TH+ neurons producing DA were obtained from SDIA-treated ESCs. When transplanted, SDIA-induced dopaminergic neurons integrated into the mouse striatum and remained positive for TH expression [17]. In accordance with Lee's group, Kawasaki et al. also avoided using RA in their experiment, as RA seemed to perturb neural patterning and neuronal identities in EBs, as a strong teratogen. Efficiency of DA neuron induction in the SDIA method is as high as maximum efficiency (~30%) obtained by Lee's method with SHH, FGF8 and ascorbate treatment [17]. Neural induction by SDIA provided a new powerful tool for both basic neuroscience research and therapeutic applications.

Low efficiency of generation of DA neurons from primary cultures of foetal neonatal cells, or adult stem cells, limits their therapeutic potential as donor cells [18]. In effort to improve efficiency of DA neuron generation, survival and maturation in vitro, Kim et al. used a cytomegalovirus plasmid (pCMV) driving expression of rat Nurr1 complementary DNA modified to establish stable Nurr1 ESC lines [18]. Nurr1 ESCs raised the proportion of TH+ neurons up to 78%, combined with their previous five-stage differentiation method [16, 18]. They also demonstrated that these DA neurons from ESCs could functionally integrate into host tissue as well as lead to recovery in a rodent model of PD [18].

These results have subsequently been replicated using hESCs with some modifications, but efficiency was not satisfactory [19-21]. Perrier et al. have reported that co-culture of hESCs on MS5 stroma can yield highly efficient differentiation into midbrain DA neurons [22]. Neural differentiation of hESCs was induced by means of co-culture on MS5, MS5-Wnt or S2 stroma at comparable efficiencies. Growth factors were added in various combinations and at various time points. Rosette structures were harvested mechanically from feeder layers on day 28 of differentiation and gently replanted on 15 g/ml polyornithine/1 g/ml laminin-coated culture dishes in N2 medium supplemented with SHH, FGF8, AA and BDNF. These cells were resuspended in N2 medium, and replated again on to polyornithine/laminin-coated culture dishes in the presence of SHH, FGF8, AA and BDNF. After additional 79 days culture, cells were found to have differentiated in the absence of SHH and FGF8 but in the presence of BDNF, glial cell line-derived neurotrophic factor, transforming growth factor type 3, dibutyryl cAMP and AA [22]. The workers observed that exposure to SHH and FGF8 from day 12 to day 20 differentiation, followed by differentiation in the presence of AA and BDNF, resulted in 3-fold increase in TH+ cells. Up to 79% of all the neurons express TH, the rate-limiting enzyme in the synthesis of DA. In addition to TH expression, cells in these cultures expressed key markers associated with normal midbrain DA neurons. However, the high-yield midbrain DA neuron derivation protocol reported here need to be transplantated into pre-clinical animal models of PD [22]. Beyond this, cell survival and long-term maintenance of phenotype are essential parameters for testing in vivo.

In 2008, a group of Korean scientists reported a method for differentiating hESCs into functional TH+ neurons, with up to near 86% total hESC-derived neurons, the highest purity ever reported [23]. The unique feature of their protocol was generation of pure spherical neural masses (SNMs). These SNMs could be expanded for long periods without losing their differentiation capability and could be coaxed into DA neurons efficiently within a relatively short time (approximately 2 weeks) when needed. SNM culture and DA neuron derivation from the SNMs did not need feeder cells, which reduced risks of contamination by unwanted cells and pathogens. More importantly, their hESC-derived DA neurons induced clear behavioural recovery after transplantation in a Parkinsonian rat model, indicating their functionality in vivo [23].

It has been reported that bone marrow mesenchymal stem cells (BMSCs) can differentiate into not only osteogenic, adipogenic, chondrogenic cells, but also into other lineages including myogenic, hepatic and neurogenic cells [24]. Furthermore, they are inducible to differentiate into cells with the DA neuronal phenotype suggested by expression of TH, DAT markers, as well as synthesis and secretion of DA after appropriate stimuli [25]. Previous studies have shown that human BMSC engraftment can alleviate motor dysfunction in Parkinsonian animal models, but with limited efficacy and but few engrafted cells surviving. Our team transplanted equal amounts of hBMSCs into hemi-lesioned Parkinsonian rats with supplementation of bFGF, to assess whether a combination of bFGF and hBMSC therapy would enhance treatment effectiveness in PD rat models [26]. As a result, bFGF promoted hBMSCs to transdifferentiate towards neural-like lineages in vitro [26]. In addition, hBMSC transplantation alleviated motor functional asymmetry, and prevented DA neurons from loss in the PD model, while bFGF administration enhanced neurodifferentiation capacity and therapeutic effect [26].

Similar strategies have been applied for differentiating hiPSCs into DA neurons. Cooper et al. postulated that a major limitation for experimental studies of current ESC/iPSC differentiation protocols, was lack of VM DA neurons of stable phenotype, as defined by expression marker code FOXA2/TH/-tubulin [27]. They demonstrated a combination of three modifications that were required to produce VM DA neurons. First, early and specific exposure to low-dose RA improved regional identity of neural progenitor cells derived from pluripotent stem cells. Secondly, a high activity form of human SHH established a sizeable FOXA2+ neural progenitor cell population in vitro. Thirdly, early exposure to FGF8a, rather than FGF8b, and WNT1 were required for robust differentiation of the FOXA2+ floor plate-like human neural progenitor cells into FOXA2+ DA neurons [27]. FOXA2+ DA neurons were also generated when this protocol was adapted to feeder-free conditions. In summary, their new human ESC and iPSC differentiation protocol can generate human VM DA neurons as required for relevant new bioassays, drug discovery and cell-based therapies for PD [27].

The majority of PD cases are sporadic with unknown cause. Age, oxidative stress, toxin and environmental factors are risk factors [2], and remaining 10% is familial PD, where several causative genes have been identified [28]. Before stem cell modelling appeared, the most used cell or animal models for PD were generated with toxins such as rotenone, 6-OHDA, MPTP or genetic models. The relationship between MPTP and PD was found in a cluster of young drug addicts, by Davis et al. in 1979 [29]. MPTP easily crosses the bloodbrain barrier (BBB) where it is oxidized in glial cells into MPP+. MPP+ competes with DA for the DA transporter and after entering neurons, it exerts its toxic effect by inactivating complex I of the ETC [30]. MPTP is commonly used to model for PD in primates and rodents in that the drug kills dopaminergic neurons, allowing researchers to study neuronal circuitry with reduced dopaminergic involvement [31]. Many workers have demonstrated that MPTP administration is able to reproduce most, but not all, the clinical and pathological hallmarks of PD in monkeys [32-34] and, at least degeneration of dopaminergic neurons, in mice [35]. Similar to MPTP, the pesticide rotenone disrupts complex I function of mitochondria [36]. Our team has demonstrated that rotenone models for PD appear to mimic most clinical features of idiopathic PD and recapitulate the slow and progressive loss of DA neurons and LB formation in the nigral-striatal system [36]. Both MPTP and rotenone have been important for establishment of PD animal models. However, while they promote dopaminergic neuron death with associated motor impairment, their side effects and lack of specificity are major drawbacks [31]. 6-OHDA, a selective catecholaminergic neurotoxin, is used to generate lesions in the nigrostriatal DA neurons in rats [37]. Unlike MPTP, 6-OHDA cannot cross the BBB. So, 6-OHDA must be injected into the SNc, medial forebrain bundle or striatum, to induce Parkinsonism, rather than systemic administration [38]. Intrastriatal injection of 6-OHDA causes progressive retrograde neuronal degeneration in the SNc and VTA [39, 40]. Genetic models provide us with better understanding of underlying genetic forms of PD, even though their pathological and behavioural phenotypes are often quite different from the human condition [41]. Many genetic variant models, including SNCA, LRRK2, PINK, Parkin, DJ-1 and Glucocerebrosidase (GBA), have been generated to explore inherent mechanisms in PD [42-45]. The well-established genetic models are able to interpret pathogenesis of only 510% familial PD, however, without replicating the entire genetic background of the patients in vitro [46]. Moreover, differences between species in displaying neurodegenerative phenotypes make it difficult to extrapolate results obtained from animal models to humans [47]. The discovery of iPSCs has for the first time enabled us to reproduce DA neurons from individuals who suffered from familial or sporadic PD [47, 48]. Moreover, these iPSC models allow us to explore pathogenic factors and discover interactions between genetic and exogenous factors involved in the pathogenesis of PD. As individuals' responses to drug compounds varies, patient-specific iPSCs may be used to distinguish between those individuals likely to respond to new therapeutics and those who are not, and more accurately predict toxicity and efficacy in screening drugs, from mechanisms, in comparison to animal models.

Differentiation of DA neurons from iPSCs has been demonstrated to be relatively robust and reproducible, allowing for generation of disease models from patients carrying a variety of mutations in key genes implicated in familial PD, including PARK2, PINK1, LRRK2, SNCA and GBA [49, 50]. Among genetic risk factors, Parkin (PARK2) is the most frequently mutated gene that has causally been linked to autosomal recessive early-onset familial PD [51]. In patients with PD onset before the age 45, PARK2 mutations are seen in up to 50% of familial cases and about 15% of sporadic cases [52]. Parkin knockout mouse models display some abnormalities, but do not fully recapitulate the pathophysiology of human PARK2. Jiang et al. generated iPSCs from normal subjects and PD patients with Parkin mutations and demonstrated that loss of Parkin in human midbrain DA neurons greatly increased expression of monoamine oxidases and oxidative stress, significantly reduced DA uptake and increased spontaneous DA release [53]. These results suggest that Parkin controls dopamine utilization in human midbrain DA neurons by enhancing the precision of DA neurotransmission and suppressing DA oxidation [53]. Imaizumi et al. used PARK2 patients' specific iPSCs-derived neurons to recapitulate pathogenic changes in the brain of PARK2 patients [54]. The data indicated that PARK2 iPSC-derived neurons exhibited increased oxidative stress, impaired mitochondrial homoeostasis and SNCA accumulation [54]. Recently, Ren et al. showed that the complexity of neuronal processes and microtubule stability were significantly reduced in iPSC-derived neurons from PD patients with Parkin mutations [55], suggesting that Parkin maintains morphological complexity of human neurons by stabilizing microtubules [55]. Shaltouki et al. observed reduced DA differentiation, accompanied by reduced mitochondrial volume ratio and abnormal mitochondrial ultrastructure, consistent with the current model of PARK2 mutations [48].

PINK1 functions upstream of Parkin, and is involved in recruiting Parkin to damaged mitochondria, for example, following mitochondrial depolarization [56]. Mutations in either PARK2 or PINK1 result in impaired mitophagy. Cooper et al. found that PINK1 mutant iPSC-derived DA neuronal cells were more sensitive to cell death and production of ROS elicited by mitochondrial and oxidative stressors, and further showed increased basal oxygen consumption and proton leakage suggestive of intrinsically damaged mitochondria [57]. Moreover, cell vulnerability associated with mitochondrial function in iPSC-derived neural cells could be rescued with coenzyme Q10, rapamycin or LRRK2 kinase inhibitor GW5074. These data demonstrate that iPSC-derived neural cells are sensitive models for measuring vulnerability and doseresponses of candidate neuroprotective molecules and might help to identify disease causes and better individualize treatment efficacy [57].

Mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with sporadic and familial forms of PD. Nguyen et al. have reported that DA neurons derived from G2019S mutation-iPSCs have high levels of expressions of key oxidative stress-response genes and SNCA protein [58]. The mutant neurons were more sensitive to caspase-3 activation and cell death caused by exposure to stress agents, such as hydrogen peroxide, MG-132 and 6-hydroxydopamine [58]. Cooper et al. indicated that LRRK2 G2019S and R1441C mutations reduced availability of substrates for oxidative phosphorylation, and were associated with disrupted mitochondrial movement in PD patient-specific iPSCs [57]. Laurie et al. further demonstrated the mechanisms by which LRRK2 mutations lead to loss of mitochondrial function. Their data revealed that mitochondrial DNA (mtDNA) damage was induced in neural cells by PD-associated mutations in LRRK2, and this phenotype could be functionally reversed or prevented by zinc finger nuclease (ZFN)-mediated genome editing in iPSCs [59]. These results indicate that mtDNA damage is likely to be a critical early event in neuronal dysfunction that leads ultimately to LRRK2-related PD [59].

The first genetic cause identified for familial PD was SNCA, as PD can be caused by mutations in SNCA or by overexpression of normal SNCA via gene duplication or triplication, consistent with a gain-of-function mechanism. iPSC-derived midbrain DA cultures from SNCA triplication patients exhibit several disease-related phenotypes in culture, including accumulation of SNCA, inherent overexpression of markers of oxidative stress and sensitivity to peroxide-induced oxidative stress [60]. iPSCs, reprogrammed from patients with the most common A53T-SNCA mutation, had high nitric oxide and 3-NT levels compared to controls [61].

GBA mutations, which cause the lysosomal storage disorder Gaucher disease, have recently been linked to a 5-fold greater risk of developing Parkinsonism than non-carrier individuals [62], and are the strongest genetic risk factor for PD known to date. GBA1 mutated iPSC-derived neurons have low glucocerebrosidase activity and protein levels, and high SNCA levels as well as autophagic and lysosomal defects [63]. Mutant neurons display dysregulation of calcium homoeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium [63]. These findings using iPSC technology, have provided evidence for a link between GBA1 mutations and complex changes in autophagic/lysosomal system and intracellular calcium homoeostasis, underlying vulnerability to neurodegeneration. As monozygotic twins share identical genetic makeup, twin studies have been valuable for dissecting complex gene-environmental interactions in PD. Woodard et al., using iPSC technology [64], investigated a unique set of monozygotic twins and found that SNCA clearance was impaired in midbrain DA neurons carrying GBA N370S regardless of disease status. Moreover, DA levels of twins discordant for PD were different, suggesting that non-genetic factors further perturbed DA homoeostasis in addition to GBA mutations. These results verified the interactions between genetic and environmental factors in the progress of PD, and offer a theoretical basis for personalized medicine in PD (Table 1).

R42P

EX3DEL R275W

More sensitive to cell death and production of ROS elicited by mitochondrial and oxidative stressors, and increased basal oxygen consumption and proton leakage

Phenotype rescue using coenzyme Q10, rapamycin or the LRRK2 kinase inhibitor GW5074

Increased expression of key oxidative stress-response genes and -synuclein

More sensitive to caspase-3 activation and cell death caused by exposure to stress agents

L444P,

N370S

Reduced glucocerebrosidase activity and protein levels, increased -synuclein levels as well as autophagic and lysosomal defects

Dysregulation of calcium homoeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium

Although the cause of sporadic PD is not fully understood, various factors including environmental toxins, genetic susceptibility and age, have been implicated. Isogenic hiPSC PD models show that toxin-induced nitrosative/oxidative stress results in S-nitrosylation of transcription factor MEF2C and this redox reaction inhibits MEF2C-PGC1 transcriptional network, contributing to mitochondrial dysfunction and apoptotic cell death [65], suggesting that the MEF2C-PGC1 pathway may be a new drug target for PD. The advance of iPSC technology now enables widespread development of PD models for dissecting molecular mechanisms that contribute to its disease pathogenesis.

As previously mentioned, there is currently no cure for PD except for some extent of relieving the symptoms. Current treatments include the use of oral medication of l-DOPA dopamine receptor agonists, MAO-B, apomorphine in more serious cases, continuous intestinal infusion of l-DOPA, and deep brain stimulation (DBS) in subthalamic nucleus and globus pallidus by using surgically implanted electrodes [66]. l-DOPA is the gold standard for treatment of PD. Up to now, no medical nor surgical therapy has been shown to provide superior anti-parkinsonian benefits than can be achieved with l-DOPA [67]. Unfortunately, its therapeutic effect is reduced after around 35 years use [68]. The problems and limitations associated with long-term use of l-DOPA, including on-off fluctuations and emergence of dyskinesia, facilitating exploration of better ways to restore dopamine neurotransmission. Dopamine receptor agonists are used as the first choice to delay initiation of l-DOPA treatment, with longer plasma elimination half-lives than l-DOPA. Their mechanism of action are by stimulation of presynaptic and postsynaptic DA receptors so that their use has therefore been considered to be opportunity to improve continuous drug delivery [67]. Selegiline was the first selective, irreversible inhibitor of monoamine oxidase type B (MAO-B) used in treatment of PD, which can stabilize DA levels in the synaptic cleft [67]. Because of its capacity for interfering with oxidative stress and for blocking MPTP toxicity, selegiline has been tested in the first major trial as a putative disease-modifying agent [67]. Rasagiline is another MAO-B inhibitor, with different metabolites than selegiline, successfully developed for PD therapy. Good tolerance to rasagiline and its ease of use make it an appealing option at the start of therapy [67]. DBS as surgical treatment has some serious limitations. It is costly and can produce cognitive disorders, which may be permanent [68]. All of these treatments have considerable side effects such as ultimate loss of drug effect (wearing off) during disease progression, occurrence of dyskinesia (notably with l-DOPA) use, and appearance of non-motor symptoms that are largely refractory to dopaminergic medication [69]. The concept of using cell transplantation to substitute for loss of DA neurons in the brains of PD patients has evolved. In addition to conventional clinical treatments, such as pharmaceutical drugs and DBS, cell replacement therapy has offered a novel basis for development of effective therapeutic strategies for PD. In 1987, Brundin et al. first transplanted human VM tissue into the striatum of PD patients in Sweden, and the era of cell therapy for PD patients started [70]. Various source tissues have been assessed for therapeutic replacement of DA neurons, such as hESC, hiPSC or DA grafts directly converted from somatic cells. Our team also has transplanted DiI-labelled human umbilical cord mesenchymal stem cells (HUMSCs) to rotenone-induced hemiparkinsonian rats [71]. We showed that intra-CPu transplantation of DiI-labelled HUMSCs 4 weeks after rotenone administration ameliorated APO-induced rotations gradually over a period of 12 months, indicating long-term therapeutic effect of this approach [71]. By monitoring red fluorescence of DiI, we found that HUMSCs migrated in the lesioned cerebral hemisphere, from CPu to SNc, or even to the opposite hemisphere through the corpus callosum. HUMSCs survived for up to 12 months after transplantation, and differentiated into Nestin-, NSE-, GFAP- and TH-positive cells in the CPu and TH+ cells in the SNc. No tumour-like structures was observed in implanted CPu [71]. As reported, vascular endothelial growth factor (VEGF) is a neurotrophic factor which has been proven to promote growth and survival of DA neurons in VM explants and animal models for PD [72-74]. Our previous work has also indicated that relatively low-level expression of VEGF in the striatum protects DA neurons of Parkinsonian rats [75]. Next, we developed a more effective neurorestorative and neuroregenerative therapy combining VEGF and HUMSC [76]. As a result, intrastriatal infusion VEGF-expressing HUMSCs to rotenone-induced Parkinsonian rats provided a significant behavioural improvement, more significant than HUMSC transplantation alone, and resulted in revival of TH immunoreactivity in the lesioned striatum and SNc [76]. Importantly, VEGF expression enhanced neuroprotective effects by promoting DA neuron-orientated differentiation of the HUMSCs. Thus, our findings have presented the suitability of HUMSC as a vector for gene therapy and suggested that stem cell engineering with VEGF may improve transplantation strategies for PD treatment [76].

iPSCs, induced pluripotent cells, have the potential capacity for self-renewal and are able to differentiate into any somatic cells, including DA neurons [77]. Alternatively, iPSCs have properties similar to ESCs but can be generated from adult human cells such as skin, adipose tissue and fibroblasts [10]. Thus, they are ethically more acceptable than some other stem cells sources. In theory, iPSCs from patients are without risk of immunological rejection for autografting [78].

Before successfully generating hiPSCs, many efforts had been made in animal experiments. In 2006, Yamanaka et al. generated iPSCs from mouse embryonic fibroblasts (MEF) and adult mouse tail-tip fibroblasts, by retrovirus-mediated transfection of four transcription factors, Oct3/4, Sox2, c-Myc and Klf4 [10]. One year later, iPSCs were derived by the same group [79] by viral reprogramming of human skin fibroblasts with the same four factors. These studies opened a new avenue for generating patient- and disease-specific pluripotent stem cells. Wernig et al. used 6-OHDA-lesioned rats to examine whether DA neurons derived from directly reprogrammed fibroblasts had therapeutic potential for PD animals [78]. As a result, in the striatum of rats grafted with differentiated iPSCs, a large number of TH+ cells with complex morphologies have been observed, these grafted stem cells were also positive for En1, VMAT2 and DAT. Four of the five transplanted animals which contained large numbers of TH+ neurons showed marked recovery of rotation behaviour 4 weeks after transplantation [78].

Kikuchi et al. first grafted hiPSC-derived DA neurons into the brains of an MPTP-lesioned Parkinsonian monkey, which survived as DA neurons as long as 6 months [80]. In order to reduce immune rejection, Deleidi et al. generated iPSCs from cynomolgus macaque (CM) skin fibroblasts carrying specific major histocompatibility complex (MHC) haplotypes, observing that neither tumour formation nor inflammatory reactions occurred in the transplanted animals, up to 6 months after transplantation [81]. Concerning the aspect of directed differentiation of iPSC into DA neurons, Snchez-Danes et al. indicated lentiviral vectors driving controlled expression of LMX1A was an efficient way to generate enriched populations of human VM DA neurons [82]. However, Mak et al. revealed that the protocol using dorsomorphin and SB431542 to replace SHH with purmorphamine or smoothened agonist could greatly improve conversion of hiPSCs to the neuronal lineage [83]. These histocompatible iPSCs may allow pre-clinical validation of safety and efficacy of iPSCs for PD.

Two commonly used anti-convulsants drugs, valproic acid (VPA) and zonisamide (ZNS), have been tested to promote differentiation of iPSC-derived DA neurons [84]. As iPSC-derived donor cells inevitably contain tumourigenic or inappropriate cells, finding better protocols to purify and sort iPSCs is urgent. Doi et al. have shown that hiPSC-derived DA progenitor cells can be efficiently isolated by cell sorting using a floor plate marker, CORIN [85]. When transplanted into 6-OHDA-lesioned rats, CORIN+ cells survived and differentiated into midbrain DA neurons in vivo, resulting in significant improvement in motor behaviour, without tumour formation [85]. Recently, Hallett et al. analysed CM iPSC-derived midbrain DA neurons for up to 2 years following autologous transplantation in a PD model. They observed that unilateral engraftment of CM-iPSCs provided gradual onset of functional motor improvement, and increased motor activity, without any need for immunosuppression. Postmortem analyses demonstrated robust survival of midbrain-like DA neurons and extensive outgrowth into the transplanted putamen [86]. These experiments offered strong immunological, functional and biological rationales for using midbrain DA neurons derived from iPSCs for future cell replacement in PD (Table 2).

Although pre-clinical studies concerning iPSCs-derived cell therapies have shown great achievement, yet, some limitations hinder clinical usage of iPSCs for PD treatment. Tumourigenicity of iPSCs is an an important putative problem. Murine iPSCs and ESCs both form teratomas when transplanted into syngeneic mice. Also, hiPSCs and hESCs generate teratomas when injected into immunodeficient mice [87]. A standardized sensitive teratoma assay to detect low numbers of tumour-forming cells within a therapeutic cell preparation would be highly valuable. Gropp et al. presented detailed characterization of an efficient, quantitative, sensitive and easy-to-perform teratoma assay [87]. These tumours may be benign (also, they may be malignant), although even so can become fatal when very large. In some studies, when teratomas have been removed from mice, the animals survived [88]. Importantly, aggressiveness of teratocarcinomas from iPSCs is greater than that of ESCs [88]. Differences in oncogenicity between ESCs and iPSCs might be due to their different approach of being derived [88]. As hiPSCs have been first derived by transduction of human dermal fibroblasts with integrating viruses carrying four transcription factors Oct4, Sox2, c-Myc and Klf4 [79], c-Myc is a well-established oncogene while the other three transcription factors are known to be highly expressed in various types of cancer [89-91]. Yamanaka et al. subsequently reported a further Myc family member, L-Myc, as well as C-Myc mutants (W136E and dN2), all of which indicated little transformational activity, promoting hiPSC generation more efficiently and specifically compared to WT C-Myc [32]. A further cause of tumourigenicity may be attributed to random integration of foreign DNA into the host genome disrupting important genes or activating oncogenes, potentially leading to uncontrollable growth of cells [92].

Stadtfeld et al. generated mouse iPSCs from fibroblasts and liver cells by using non-integrating adenoviruses transiently expressing Oct4, Sox2, Klf4 and c-Myc [93]. These adenoviral iPSCs (adeno-iPSCs) showed DNA demethylation characteristic of reprogrammed cells, expressed endogenous pluripotency genes and formed teratomas [93]. Their work indicated that insertional mutagenesis was not required for in vitro reprogramming [93]. Thus, more than 2 years after establishment of iPSC technology by Yamanaka's group, these newly generated adeno-iPSCs have been the first reported reprogrammed pluripotent stem cells with evidence of complete lack of viral transgene integration [94]. Yamanaka et al. described an alternative method to generate iPSCs from MEFs by continual transfection of plasmid vectors free from plasmid integration [95]; this protocol took around 2 months to complete, from MEF isolation to iPSC establishment. The virus-free technique reduced safety concerns for iPSC generation and application, and have provided a source of cells for investigation of mechanisms underlying reprogramming and pluripotency [95]. Introducing mRNA directly into host cells without altering their genomic makeup or using episomal DNA-based vectors which seldom integrate into the host genome, holds the potential to solve this problem, by providing sufficient reprogramming factor expression for successful transformation of somatic cells [96-98].

Anokye-Danso et al. showed that expression of the miR302/367 cluster rapidly and efficiently reprogramed mouse and human somatic cells to an iPSC state without requirement for exogenous transcription factors [99]. This miRNA-based reprogramming approach was two orders of magnitude more efficient than standard Oct4/Sox2/Klf4/Myc-mediated methods, and the miR302/367 iPSCs displayed characteristics similar to the Oct4/Sox2/Klf4/Myc-iPSCs [99].

A further problem which hinders iPSCs treatment is that the therapeutic effect can be influenced by inherent pathogenic features of PD. Interactions between genetic and exogenous factors result in its pathogenesis. A general concern about use of autologous iPSC transplantation is whether underlying PD-associated genetic mutations presented in transplanted neurons increases vulnerability of iPSC-derived midbrain DA neurons to disease pathology. Environmental factors and age contribute largely to the pathogenesis of PD. Thus, iPSC-derived neurons represent a reasonable strategy for more advantages. It has been shown that LBs, the pathological features of PD, can be found in grafts of foetal VM tissue [100, 101]. The first reason is that LB pathology is a reaction to inflammation from host brain tissues, possibly mediated by cell stress induced by reactive microglia [102]. Second, the spread of SNCA into the graft from the host may contribute to these pathological changes [103, 104]. However, Barker et al. believe that these pathological changes are not likely to limit widespread adoption of cell treatments, as numbers of cells with LB-like pathology in the grafts were small compared to numbers of healthy cells [105-107]. Patients can still be functionally stable, more than a decade after such a graft, at a time when accumulation of SNCA had been observed [108, 109].

Apart from the potential risk of tumourigenicity or inherent pathogenic features deriving from donor cells, pluripotent stem cell-derived cell populations for therapies also confer a risk for the contamination of transplantation cell populations with residual pluripotent cells [110]. In order to resolve this issue, several sorting methods have been developed for enrichment of differentiated neural cell populations and elimination of pluripotent stem cells, using flow cytometric analysis (FACS) or MACS [111]. Different combinations of CD markers have been explored to purify heterogeneous pluripotent stem cell-derived neural cell populations. Pruszak et al. identified a cluster of differentiation (CD) surface antigen code for the neural lineage, based on combinatorial FACS of three distinct populations derived from hESCs: combinatorial CD15/CD24/CD29 marker profiles [112]. They found that CD15(+)/CD29(HI)/CD24(LO) surface antigen expression defined NSCs, and this could eliminate tumour formation in vivo, resulting in pure neuronal grafts [112]. Yuan et al. performed an unbiased FACS- and image-based immunophenotyping analysis using 190 antibodies to cell surface markers on pluripotent stem cells [113]. From this analysis they isolated a population of NSC that were CD184(+)/CD271(-)/CD44(-)/CD24(+) from neural induction cultures of hESCs and hiPSCs [113]. To improve the sorting method, Sundberg et al. sorted primate iPSC-derived neural cell population with NCAM+/CD29low selection [111]. They demonstrated that teh NCAM+/CD29low selection method enriched the FOXA2/TH and EN1/TH+ DA neurons in vitro compared to unsorted cell populations from >10% prior to sorting to > 35% after sorting. Importantly, sorting with NCAM+/CD29low selection prior to transplantation eliminated non-neural tumourigenic cells from the grafts and significantly increased the number of TH+ cells in the cell grafts compared to unsorted cell populations [111].

In this review, we have summarized a number of scientific and ethical issues in modelling and treatment with iPSCs for PD. iPSCs can be employed as relevant Parkinsonian cell models, for drug screening, studying disease progression and most importantly for treatment of PD by transplantation techniques. Compared with other stem cells, iPSCs stand out for their powerful pluripotency, few ethical issues and less immune rejection, although there are still several issues that need to be solved prior to translation of iPSCs into the clinical setting. First, the exact mechanisms of how transplanted cells restore host brain function and how to connect them with circumjacent brain tissues have not been yet elucidated. Second, tumourigenicity of iPSCs may surpass their therapeutic effects. Ultimately, iPSC, derived from autologous PD patients, may contain pathogenic gene mutations that affect prognosis of transplantation therapy. With improvements in differentiation methodologies and better understanding of pathogenesis of PD through patient-specific iPSCs, iPSC therapy can be a potential alternative for PD treatment combined with traditional drug development platforms and gene therapy.

This work was supported by grants 31171211 and 81471305 from the National Natural Science Foundation of China (to TW), grant 81200983 from the National Natural Science Foundation of China (to NX), grant 81301082 from the National Natural Science Foundation of China (to JSH), grant 2012B09 from China Medical Foundation (to NX) and grant 0203201343 from Hubei Molecular Imaging Key Laboratory (to NX). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

There are no actual or potential conflicts of interest.

2016 John Wiley & Sons Ltd

Request Permissions

Please enable Javascript to view the related content of this article.

Advertisement

Follow this link:
Induced pluripotent stem cells and Parkinson's disease ...