Category Archives: Stem Cell Medicine

Prof Ziad Mallat leads Cambridge effort to win 30m to tackle leading cause of heart attacks and strokes – Cambridge Independent

It is the worlds biggest killer and yet we dont fully understand the leading cause behind it.

Cardiovascular diseases claimed an estimated 17.9 million lives in 2016 31 per cent of all deaths around the globe.

And 85 per cent of these were due to heart attacks and stroke, most commonly caused by a blockage of the arteries known as atherosclerosis.

Now an international team led by a Cambridge professor of cardiovascular medicine is competing for a 30million prize from the British Heart Foundation to unravel its secrets.

If they beat the other three shortlisted teams in the charitys Big Beat Challenge, they will create the worlds first 3D map of atherosclerosis at single cell resolution, giving unparalleled insight into this hardening or blocking of the arteries.

Prof Ziad Mallat, of the Department of Medicine at the University of Cambridge, tells the Cambridge Independent:We are excited about the prospect of this. We hope we have assembled the right team.

Atherosclerosis is very debilitating. If it happens in the arteries that supply the brain, it causes stroke. If it happens in the arteries supplying the heart, it causes heart attacks.

It is really common across the world. Every five minutes in the UK there is one heart attack and one stroke.

Why is this having such a huge impact on the quality of life of people? We believe something is not being treated or understood.

Clinicians currently treat the risk factors for the disease, which include high blood cholesterol, high blood pressure and diabetes.

What we dont do is really treat what causes the disease, which is the malfunctioning of the immune system, says Prof Mallat.

When you have high blood pressure or cholesterol, this injures the arteries. Initially, the immune system sends immune cells to the injured vessel to try to heal the artery.

However, what we know is that most of the time the immune system doesnt operate properly and this prevents the healing, and so the disease progresses.

We have good understanding of how this happens in pre-clinical models, like mouse models, but very limited understanding of how it happens in humans.

We think this is what is preventing doctors and scientists from finding a treatment that would transform the way patients are treated.

Through their iMap, as they are calling it, Prof Mallat and the team of global experts he has assembled want to understand what is happening in the accumulations, known as plaques, that block the arteries and affect blood flow to the heart and other parts of the body. The plaques can be made up of fat, cholesterol, calcium and other substances.

These plaques obstruct the lumen [the interior space in the artery] and even burst into the lumen, leading to clot formation, which obstructs the blood flow. This causes the heart attacks and strokes, says Prof Mallat.

Our idea is to build the first 3D map of these fatty plaques, at

. We would like to know what each immune cell and each cell in the vessel wall is doing. What is its genetic make-up? What is its protein make-up? What is the fuel that it is using? Why, when the immune cell comes along to do a good job, does it stop doing it?

We want to interrogate each cell and work out how it is interacting and communicating with other cells.

Only with this 3D map of the plaques will we be able to understand what is happening inside. Once we have done this, we will be able to harness this knowledge to find new protective methodologies and therapies.

These therapies could harness the immune system, which raises the possibility of vaccinating against atherosclerosis.

If we understand how the immune cells react, we can use the information to re-educate them with vaccination, suggests Prof Mallat. If they are overreacting to fat components or protein components, we can educate them to make them do the right job when they see this in the arteries, to reduce the inflammation and limit the development of the disease.

The scale of this challenge, however, is vast and requires a multi-disciplinary approach.

It needs a lot of different expertise around the world, says Prof Mallat. You need good cardiologists, good molecular biologists, immunologists, mathematicians and computer scientists because the information will be huge and needs to be integrated together. You need people who know a lot about genomics, lipidomics and proteomics, so we have gathered world-leading experts in each of these areas to come together and look at this problem from every angle possible.

Among those helping Prof Mallat is Sarah Teichmann, from the Wellcome Sanger Institute at Hinxton, who is the co-founder of the global consortium working on the Human Cell Atlas a hugely ambitious and important project creating comprehensive reference maps of all human cells in the human body.

They are looking at the make-up of healthy organs, notes Prof Mallat. Some of the investigators are mapping some of the arteries and are looking at vascular cells like endothelial cells. It is intriguing but nobody else is looking at other cells in the artery. We are looking at both the healthy arteries and the diseased arteries. It is building on the work of the Human Cell Atlas.

Also on the team are experts from Imperial College London, Germany, France, Spain, the La Jolla Institute of Immunology in San Diego and from Icahn School of Medicine at Mount Sinai in New York.

Key to their work is the need for data and samples, and the group has multiple sources available.

We have organ donors from the Cambridge bio-repository and the clinical school at Mount Sinai, so we have access to healthy and diseased arteries from the same individuals.

We have access to blood from these individuals and to immune cells from other parts of the body, so we can compare what the immune cells are doing in different compartments.

The other source is from a cohort of thousands of individuals, through a collaboration with Professor Valentin Fuster in Madrid, who have been followed for more than 10 years, and they will be followed for another 10 years.

We have blood samples and microbiota from them. We also have access to imaging of their arteries. They are followed for cardiovascular outcomes, so if someone has a heart attack or stroke, it is documented.

We will be able to look at the ageing of the immune system in these individuals and how this correlates to changes in their arteries and the occurrence of disease.

All of this is being done at very high resolution, which has not been done before. Integrating the information from the genes, the proteins, the lipids and so on, to have a broad view, has never been possible.

There are parallels with the work being carried out at Cancer Research UK Cambridge Institute under Prof Greg Hannon, where the first virtual 3D tumour is being created using a multi-disciplinary team.

We are discussing with him how we can integrate some of the technologies he is developing. It will be fantastic to collaborate with him on this, says Prof Mallat.

What is known already is that our arteries are sensitive to changes in blood flow.

Even subtle perturbations in the micro-environment are sensed by the arteries and can be considered as a danger, explains Prof Mallat.

When it interprets this as a danger, it sends signals to the immune system to react. I would say this is happening almost continuously, and is aggravated of course when you have additional stimuli like high blood cholesterol or exposure to smoke.

While the use of imaging and monitoring of biomarkers is helping us diagnose atherosclerosis earlier, Prof Mallat describes this as not optimal, because we dont understand the disease in a comprehensive manner. A 3D map would aid diagnosis, prediction and prevention of disease, as well as opening up new therapeutic avenues.

Nobody knew 10 or 15 years ago that the immune system could play such a huge role in cancer, Prof Mallat points out. Now cancer immunotherapy is advancing enormously. We are convinced that atherosclerosis is highly motivated by the immune system but no-one is targeting the immune system to treat it. Thats why we want to understand it and we think this could really induce a revolution in our understanding and how we treat it.

Cambridge Cardiovascular to host events at Cambridge Science Festival

Visitors to Cambridge Science Festival will have a chance to find out more about the iMap project and the work of cardiovascular researchers.

Cambridge Cardiovascular, an umbrella group for the field, is involved in organising activities once again at this years festival, which runs from March 9 to 22.

At 6-7pm on Wednesday, March 18 at the Mill Lane lecture rooms in Cambridge, a talk titled More than a blocked pipe: The hardening of the arteries and their role in stroke and heart attacks will be delivered by Dr Nick Evans, of the Department of Medicine, and Prof Melinda Duer, of the Department of Chemistry.

At 6-7pm on Friday, March 20, also at Mill Lane lecture rooms, Dr Sanjay Sinha, of Cambridge Stem Cell Instituteand the Department of Medicine will discuss Mending broken hearts: stem cells for heart disease.

Then, from 11am to 4pm on Sunday, March 22, A View of the Heart will be on offer at the Cambridge Academy for Science and Technology, in Long Road, where cardiovascular scientists will help you explore the organ and visualise heartbeats.

Book at sciencefestival.cam.ac.uk.

The Big Beat Challenge

The British Heart Foundations 30million Big Beat Challenge is designed as the charitys moon-shot to propel our understanding of cardiovascular disease into a new era.

Some 75 applications were received from 40 countries following its launch in August 2018, and these have been whittled down to four, including the one led by Prof Mallat to map and treat atherosclerosis. The other ideas are:

Hybrid heart

Led by Jolanda Kluin, professor of translational cardiothoracic surgery at the University of Amsterdam in the Netherlands, this team plans to create a solution for heart failure by developing a soft robotic heart. They intend to design, build, test and implant a hybrid heart that consists of a soft robotic shell forming the soft artificial muscles and sensors to enable natural motion, and a tissue-engineered lining to make sure all the surfaces in contact with blood are safe. With wireless energy transfer, the vision is that this could replace the need for human heart transplantation.

Echoes

Led by Professor Frank Rademakers, chief medical technology officer at University Hospitals Leuven, Belgium, this team would develop wearable technology that can be used in daily life to capture more data than ever before. This information ranging from symptoms and physical activity to heart function and air quality could be used alongside genetic and healthcare data to transform diagnosis, monitoring and treatment of heart and circulatory diseases through the creation of a digital twin.

Cure heart

This project aims to provide a cure for inherited, killer heart muscle diseases. Led by Professor Hugh Watkins, BHF chair of cardiovascular medicine at the University of Oxford, these researchers will develop a treatment that targets and silences the faulty genes responsible for cardiomyopathies diseases of the heart muscle that can lead to sudden death at an early age. They intend to combine a deep understanding of underlying genetic mechanisms with new technologies, to stop the progression of the damage caused by genetic heart muscle diseases, or even reverse the damage.

Professor Sir Nilesh Samani, medical director at the British Heart Foundation, said: Heart and circulatory diseases remain the number one cause of death worldwide.

Were taking small steps forward every year but whats needed is a giant leap, which wont be achieved by a business-as-usual approach.

The Big Beat Challenge embodies our ambition to turbo-charge progress and could lead to its own man on the moon moment. I have absolutely no doubt the winning idea will define the decade in their area.

The teams will prepare their final applications by June 14, with interviews in early September and a decision expected by the end of the year.

Read more

Our guide to the Cambridge Science Festival 2020

Can HIV be cured? Evelyn Trust-funded research at University of Cambridge probes viral latency

Inside the Cambridge lab in pole position to create a new coronavirus vaccine

Sanger Institute scientist helps unveil blueprint for extraordinary Human Cell Atlas

Prof Greg Hannon on taking over at the Cancer Research UK Cambridge Institute and creating the worlds first virtual reality tumour

Read more from the original source:
Prof Ziad Mallat leads Cambridge effort to win 30m to tackle leading cause of heart attacks and strokes - Cambridge Independent

Aspiration-assisted bioprinting for precise positioning of biologics – Science Advances

INTRODUCTION

Aggregated cells have been formed into spheroids (13), honeycombs (4), and strands (5) from a variety of different cell types and their cocultures. They have many advantages including the cellular capability to secrete extracellular matrix (ECM) components with an effective communication between cells in a native-like microenvironment (1). When cells are grown in an isolated fashion (i.e., cells in monolayers or cell-laden hydrogels), they usually do not facilitate native-like tissue microenvironment because of limited cell-cell and cell-ECM interactions (6). Three-dimensional (3D) cell aggregates, particularly tissue spheroids, are excellent candidates to mimic in vivo tissue microenvironments, which can be reconstituted to form reproducible complex tissues [such as bone (7) and pancreas (8)] and or tissue models [cancer (9)]. Furthermore, cocultured aggregates can be used as building blocks for fabricating scalable tissue complexes (2). Spheroids loaded with endothelial cells can also facilitate a denser tissue microenvironment, inherent ECM secretion, and prevascularized network (10). As a result, large-scale vascularized tissue complexes can be biofabricated using prevascularized cell aggregates as fundamental building blocks (7, 11), which allow more accurate representations of native tissues. Moreover, these tissue complexes stand to develop physiologically correct models for drug screening, disease modeling (i.e., cancer), and high-throughput screening (12). Therefore, various advantages of tissue spheroids make them a great candidate as building blocks for 3D bioprinting.

Despite these advantages, only a few methods including extrusion-based bioprinting (2, 11, 13, 14), droplet-based bioprinting (15), Kenzan (16), and biogripper approaches (4, 17) have been demonstrated for 3D bioprinting of spheroids made of cells, such as but not limited to human articular or nasal chondrocytes, human umbilical vein endothelial cells (HUVECs), human umbilical vein smooth muscle cells, etc. Extrusion-based bioprinting technique dispenses similar-sized spheroids suspended in a hydrogel ink through a glass nozzle. During bioprinting, spheroids in the hydrogel ink may self-aggregate inside the nozzle, leading to clogging issues (2). Although extrusion-based bioprinting of cellular aggregates in the form of strands can generate scalable tissues (3, 5), their use in high-precision applications, such as organ-on-a-chip platforms or microphysiological systems, is quite challenging. In droplet-based bioprinting, a single spheroid is loaded into a droplet during bioprinting, which enables the positioning of spheroids in 2D (15). Kenzan method uses an array of needles on which spheroids are skewered by a robotic arm; however, this method requires same-size spheroids since bioprinted spheroids have to fit properly in the needle array and smaller spheroids would be prone to fragment during insertion on the needle (16). Recently, the biogripper technique was introduced to manipulate defined micromolded tissue blocks. However, this technique enables bioprinting of only molded tissue blocks in the range of 600 m to 3.4 mm, where submillimeter scale is quite large for typical tissue building blocks (4, 17). All the existing techniques used in the literature suffer from the positional accuracy and precision of spheroid bioprinting; may induce substantial damage to biological, structural, and mechanical properties of spheroids; and are limited to the use of same-size spheroids for successful bioprinting.

Here, we present a new hybrid bioprinting approach through harnessing the power of aspiration forces, which enables us to pick and precisely bioprint a wide of range of biologics, with dimensions in an order-of-magnitude range (80 to 800 m), into/onto a gel substrate with minimal cellular damage. To better understand the response of biologics to the bioprinting process, we unveil the underlying mechanism explaining the physical behavior of viscoelastic spheroids and their interactions with physical governing forces during aspiration, lifting, and bioprinting. Compared to the abovementioned methods, the presented approach facilitated the bioprinting of spheroids in higher positional precision and accuracy, ~11 and ~15% with respect to the spheroid size, respectively. In addition, it enabled bioprinting in 3D with flexibility of bioprinting of nonuniform, any-size spheroids into (i) a functional gel as a scaffold-based approach or (ii) a sacrificial gel as a scaffold-free approach (Fig. 1). The newly developed aspiration-assisted bioprinting (AAB) platform, modified from a MakerBot Replicator 1 (<$1000) (see fig. S1), operates a custom-made glass pipette, which is used to pick up biologics and 3D bioprint them into or onto a gel substrate. AAB was coupled with microvalves for droplet-based bioprinting of functional or sacrificial hydrogels. To bioprint spheroids into a functional hydrogel (i.e., fibrin), fibrinogen and thrombin layers were printed via microvalve bioprinting to obtain fibrin constructs. Spheroids were then bioprinted into desired positions before the constructs were fully cross-linked. To bioprint spheroids onto a sacrificial hydrogel (i.e., alginate), microvalve bioprinting and aerosol cross-linking processes were used. The first step included the generation of sodium alginate droplets on a glass substrate. Second, calcium chloride (CaCl2) was fumed for instant cross-linking (18). Spheroids were then picked and 3D bioprinted onto the partially cross-linked alginate. Next, the print area was overlaid with sodium alginate droplets. Then, aerosol form of CaCl2 was applied again. The process was then repeated as many times as needed for building other layers. Next, bioprinted spheroids were maintained in the support gel temporally until partial fusion was realized. Last, the support gel was gently removed through decross-linking using a lyase solution.

In step 1, spheroids are picked from the cell media by a glass pipette, where required back pressure is set to lift spheroids. Afterward, spheroids can be bioprinted into sacrificial hydrogels (scaffold-free bioprinting) or functional hydrogels (scaffold-based bioprinting). In this regard, in step 2, microvalve bioprinting is used to bioprint a gel substrate, which can then be partially cross-linked using various different cross-linking schemessuch as but not limited to enzymatic, photo, and ionic cross-linkingas highlighted in step 3. Next, in step 4, spheroids are bioprinted precisely into designed positions, and spheroid bioprinting is repeated as many times as needed. Steps 2 to 4 can be repeated as needed. In step 5, bioprinted tissues are isolated from the support hydrogel (for scaffold-free bioprinting) or further grown in the functional hydrogel (for scaffold-based bioprinting). UV, ultraviolet.

The presented approach paves the way for bioprinting several tissue types and a wide range of spheroids in 2D and 3D in gel substrates with multiple applications presented throughout this paper, including (i) the development of physiologically relevant culture environments to demonstrate the collective angiogenic sprouting behavior of spheroids in a scaffold medium and (ii) fabrication of osteogenic tissues to decode the role of midterm osteogenic induction of stem cellbased spheroids (before bioprinting) on the mineralization and assembly behavior in a scaffold-free environment. In addition to spheroids, other living cells and tissue building blocks, including electrocytes from electric eel and tissue strands, can be bioprinted for a wide variety of applications, such as but not limited to tissue engineering, regenerative medicine, drug testing and pharmaceutics, disease modeling, microphysiological systems, biophysics, and biocomputing.

In the AAB process, the first step is to pick a spheroid (fig. S2) and then lift it and drag it rapidly outside the culture media by aspiration forces as shown in Fig. 2A. The spheroid should be captured with a minimum aspiration force to transfer but not break or damage it. During aspiration under an applied stress (), a spheroid with viscoelastic properties exhibits both viscous and elastic properties (19). It exhibits elastic properties like a solid at times shorter than relaxation time and like a fluid for t > , which can be described by the Maxwell modelddt+=ddt(1)where is the strain and is the modulus of elasticity of spheroids. During this process, two major impediments were experienced. First, spheroids were prone to fragment because of low cellular cohesivity and elasticity even under very low pressure levels. Second, detachment of spheroids from the pipette tip at the three-phase contact (air-liquid-tissue) was observed during lifting. The former can be mitigated by engineering of spheroids to attain better elastic properties [such as culturing them longer (20) or incorporating cells expressing higher cell-cell and cell-matrix adhesion or ECM proteins (21)], and the latter can be addressed by determining the minimum aspiration pressure (critical lifting pressure). A spheroid is lifted by pipette aspiration after overcoming gravity, buoyance force, hydraulic drag, and thermodynamic barrier at the interface. The main difficulty in lifting a spheroid is the binding energy caused by the surface tension at the interface between the culture media and air. The spheroid contributes to the lifting barrier in the form of a contact angle at the three-phase (tissue, air, and media) contact line. The energy barrier to lift a spheroid into air can be expressed asE=As1,2cos2d2(2)where As is the surface area of a spheroid, 1,2 is the surface tension between the media and air, and d is the contact angle at the three-phase contact line. During lifting, the three-phase contact angle (dynamic contact angle) is greater than the static contact angle because of dynamics effect (22). The dynamic contact angles of spheroids made of several types of cells were enumerated in table S1. By comparison, the thermodynamics barrier is about 500 times of gravity, 100 times of Stokes drag, and 5 times of Young-Dupre critical pressure. During lifting, the energy barrier can be estimated by adding a geometrical correction factor (m) as a result of slight changes in the spheroid geometry due to the aspiration of a tongue (see Fig. 2B). The initial contact length of the tongue (ht=0) of a spheroid ranges between Rp and 2Rp. Thus, m can be defined as followsm=14Rp24RS24Rp2=1Rp2RS2Rp21(RpRs)2(3)

In Eq. 3, Rp is the pipette tip radius and Rs is the spheroid radius. m can be approximated to 1 when the spheroid radius is much larger than the radius of the pipette tip. However, when lifting a small spheroid or when the radius of the pipette is comparable to that of the spheroid, m becomes much smaller. Consequently, the critical lifting pressure of a spheroid (PC) from the media can be expressed as follows (22)PC=mFmax/Ah=2mRS1,2cos2d2/(Rp2)(4)

The critical lifting pressure is proportional to the surface tension coefficient of the media-air interface, the radius of the spheroid, the dynamic contact angle at the three-phase contact line, and the tapered angle of the pipette (influences Rp, but this parameter was fixed).

(A) Time-lapse images during spheroid lifting process (at the interface of cell media and air). (B) A schematic showing physical parameters involved in lifting of a spheroid from the cell media. (C) SEM images, (D) surface tension (n = 5), and (E) the normalized collagen content of HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids (compared to HUVEC spheroids) at day 2 (n = 4; *P < 0.05, **P < 0.01, and ***P < 0.001). (F) Critical lifting pressure to lift spheroids (in the range of 200 to 600 m in diameter) (n = 5). The experimental data spread under the theoretical curve, which was determined using the experimental data for 4T1 spheroids with parameters (d = 64 and s1,2 = 57.4 mN/m). Spheroids made of other cell types had lower theoretical critical lifting pressure values (data are not shown in the paper). For instance, the theoretical critical lifting pressure for HUVEC spheroids was 20% smaller than that of 4T1 spheroids. (G) Viscoelastic behavior of spheroids under aspiration (n = 3). Here, h denotes the advancement of spheroids inside a glass pipette. The aspiration experiment used a similar pipette as that in bioprinting. The aspiration pressure was determined according to the size of spheroids satisfying the condition that spheroids could be lifted from the cell media.

To demonstrate picking and lifting of spheroids, we fabricated a wide range of spheroid types with different viscoelastic and surface tension properties using HUVECs, mouse fibroblast cell line (3T3), mouse mammary carcinoma line (4T1), human dermal fibroblasts (HDFs), coculture of human mesenchymal stem cells (MSCs) and HUVECs, and MSCs. Various kinds of spheroids in a diameter range of 200 to 600 m were obtained using cell-repellent 96-well plates. Spheroids were fabricated at a cell density from 2500 to 10,000 per spheroid and cultured for 1 to 3 days (fig. S3A). During the first 24 hours, we closely monitored spheroid formation and observed that 3T3 and HDF spheroids became compact and withstood aspiration forces at the end of 20 hours, while others became compact at 24 hours, implying that these spheroids could be lifted anytime thereafter (fig. S4). To investigate the ultrastructure of spheroids, spheroids were analyzed by scanning electron microscopy (SEM), and SEM images demonstrated clear differences in surface morphology and compactness of different spheroids cultured for 2 days (Fig. 2C). The surface topology of MSC spheroids displayed a substantial amount of secreted ECM with smoother and more compact structure due to tight cell-cell and cell-ECM interactions. On the other hand, the surface topology of HUVEC spheroids displayed lesser ECM compound, which resulted in a pitted surface and tiny gaps between cells. To lift spheroids, spheroids were required to be compact and captured in the cell media and to withstand aspiration forces. Spheroids also have surface tension, with each cell being analogous to a liquid molecule (23, 24). Thus, surface tension of spheroids were investigated by a micropipette aspiration technique (19, 25) according to the Young-Laplace equation, which gave the relationship between the internal pressure of spheroids and cell media across a curved interface. According to the Young-Laplace equation, the surface tension coefficient can be estimated as =P2(1/Rp1/Rs), where P is the equilibrium aspiration pressure when the advancement of the spheroid inside the pipette was the same as the radius of the pipette tip. As shown in Fig. 2D, the surface tension of HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids at 2 days of culture were measured to be ~14, 30, 37, 41, 51, and 66 mN/m, respectively. Surface tension of MSC-only spheroids was approximately five times higher than that of HUVEC spheroids. It is worth mentioning that Norotte et al. (26) used a different method to measure the surface tension of spheroids and obtained a similar result for HUVEC spheroids. While the surface tension of 4T1, HDF, and MSC/HUVEC spheroids increased over time, the surface tension of spheroids made from other cell types maintained similar properties during the 3-day culture (fig. S3B). This time frame, on the other hand, can vary for spheroids prepared using other techniques, such as but not limited to handing drop, microfluidic, rotator flask, and liquid overlay methods (27). Our results indicate that there is a positive correlation between the surface tension of spheroids and their compactness, confirming previous findings by Foty and Steinberg (28). In addition, the total collagen amount of spheroids was investigated using a hydroxyproline colorimetric assay kit since collagen is one of the major ECM components of spheroids (3). The measured collagen amount for 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids at day 2 was ~1.4-, 1.4-, 1.5-, 2.8-, and 3.4-fold higher compared to that for HUVEC spheroids, respectively (Fig. 2E). The normalized collagen content in different spheroids showed a similar trend as compared to the compactness of spheroids; however, we did not observe any notable differences among the normalized collagen content for 3T3, 4T1, and HDF spheroids. A recent study reported a positive correlation between compactness of cell aggregates and their collagen expression (29).

Figure 2F denotes a linear relationship between the critical lifting pressure and diameter of spheroids (in the range of 200 to 600 m). When the diameter of spheroids increased, the critical lifting pressure also increased. The theoretical critical lifting pressure for each spheroid types at different diameters were determined using Eq. 4, where the maximum theoretical value belonged to 4T1 spheroids, which could be used as a baseline for other spheroid types to be tested during aspiration. In addition, the minimum critical lifting pressure among all spheroids types (from 200 to 600 m) was determined to be ~19 mmHg. The difference between the theoretical and experimental value of the critical lifting pressure was due to (i) the irregular shape of spheroids (elliptic or pancake-like) influencing the maximum energy barrier (30, 31) and (ii) the measurement inaccuracy of the pressure sensor (which was around 10%; see fig. S5, A and B). Another vital factor for successful bioprinting was to determine viscoelastic properties of spheroids, which were reflected by the relationship between stress and strain as a function of time. During the AAB process, spheroids were kept within a solid elastic regime. The operation time of AAB was defined by the elapsed time between spheroid picking and bioprinting, which was commonly less than 30 s. Therefore, spheroids were successfully bioprinted, while their relaxation time () was longer than the bioprinting operation time. For this reason, we plotted the aspiration curves of various spheroids (Fig. 2G). At the early stage of aspiration, spheroids deformed very quickly within the elastic regime. Then, after a relaxation time, accumulated stress was gradually released, and spheroids experienced a relaxation process and return to their original shapes. As shown in Fig. 2G, the relaxation time for HUVEC, 4T1, and MSC/HUVEC spheroids were around 3 to 5 min. The relaxation time for HDF spheroids could reach up to 18 min. Overall, these relaxation times were sufficient to assure successful bioprinting without permanent deformation to spheroids.

After lifting spheroids from the cell media and transferring them to the bioprinting stage, the back pressure was released, and the spheroids were fully recovered or, in other words, they returned to their original shapes as bioprinting time was less than the relaxation time. To bioprint a spheroid, the spheroid was partially submerged into a precross-linked or partially cross-linked gel substrate, as going deeper could result in their breakage due to penetration of the pipette tip into the spheroid. When the nozzle moved up, the spheroid was stuck to the gel because of adherence between the spheroid and the gel (Fig. 3A1), released from the pipette, and deposited into the gel. In AAB, successful bioprinting was determined by a constant adherence at the pipette tip (upward, Fup) and a variable bioprinting force at the interface of gel (downward, Fdown). To determine Fup, two angles were defined: 1=arccosRpRs, and 2 is the pipette taper angle. Fup can be expressed as Fup = 2Rps cos (1 + 2), where s is the surface tension of the spheroid. At the bottom of the spheroid, the maximum force pulling the spheroid downward due to the surface tension of the gel substrate, i.e., the maximum bioprinting force provided by the gel, can be expressed as Fdown=2Rsgcos2(12). Here, g is the surface tension of the gel, and 1 is the effective angle of wetting. For successful bioprinting, the maximum force pulling the spheroid downward needs to overcome the surface tension adherence at the tip of the pipette. Thus, successful printing, as shown in Fig. 3A2, needs to satisfy the condition Fdown > Fup. However, if a spheroid is not fully recovered (when the spheroid is submerged rapidly into the gel), there can be an extra stress (Fext) at the pipette tip. The extra stress may increase the difficulty of bioprinting since Fdown needs to overcome Fext + Fup.

(A1) A schematic showing critical parameters during bioprinting. (A2) An image from the traveling camera showing spheroid placement onto a gel substrate. Fluorescent images showing (A3) PSU and (A4) matrix patterns. GFP+ MSC spheroids were patterned onto (A5) COL I and (A6) gelatin-methacryloyl (GelMA). (A7 and A8) Images showing that eight fixed spheroids were bioprinted on top of each other in air without any gel support during bioprinting. DAPI, 4,6-diamidino-2-phenylindole. (B1) Time-lapse images of self-assembly process after bioprinting of 1-day cultured 3T3 spheroids at 0, 24, 48, and 72 hours (B2) and the normalized contact length and intersphere angle of fusing spheroids up to 24 hours. (B3) Cell viability of 3T3 spheroids that were not treated with bioprinting (control), after bioprinting inside the cell media (case 1), and after bioprinting into a gel substrate (case 2) (n = 3; *P < 0.05). CMTMR, 5-(and-6)-(((4-chloromethyl)benzoyl)amino) tetramethylrhodamine; CMFDA, 5-chloromethylfluorescein diacetate. (C1) Hematoxylin and eosin staining from the sagittal plane of a tail segment of electric fish showing stacked electrocytes in series. (C2) The SEM image of a single electrocyte. (C3) Calcein staining of bioprinted electrocytes. (D) A bioprinted cartilage tissue strand between pins. Photo credit: Bugra Ayan, Penn State University.

To verify the presented theoretical approach, we measured 1, 2, and 1 angles and obtained the surface tension of precross-linked gel [1% (w/v) alginate] from the literature (32). The results indicated that Fdown was ~32, 14, 12, 11, 9, and 7 times greater than Fup for HUVEC, 3T3, 4T1, HDF, MSC/HUVEC, and MSC spheroids, respectively, as enumerated in table S2. Surface tension of gel solutions can vary from gel to gel. In some gels [such as alginate (32)], surface tension is dependent on concentration; however, there are hydrogels where surface tension is not dependent on concentration, such as polyacrylamide (33). Therefore, we cannot make a general statement on the correlation between concentration and surface tension. In addition, concentration of the gel should also be compatible with microvalve bioprinting, as higher concentrations of precursor solutions are, in general, bioprintable as such concentrations can support droplet formation (34). For bioprinting into a gel substrate, the gel precursor solution should be flowable as well so that the solution can overlay the bioprinted spheroid quickly; therefore, we performed bioprinting on partially cross-linked or uncross-linked gel precursor.

Before performing the patterning and 3D bioprinting of spheroids, we first inspected the quality of positioning during bioprinting. In this regard, agarose beads (with near-perfect sphericity) and the motion stage of the 3D bioprinter were used as control groups. The positional precision and accuracy for the motion stage (empty pipettes), spherical agarose beads (ranging from 250 to 300 m in diameter), and 2-day cultured MSC/HUVEC spheroids (~300 m in diameter) were determined to be ~2 and 2.1 m, 14.4 and 10.4 m, and 34.7 and 45.9 m, respectively (fig. S5, C to E). The results indicate that the bioprinter motion stage had a small effect on the positional error during bioprinting and that bioprinting of spheroids was about two- and fourfold less precise and accurate compared to printing of agarose beads, respectively. This could be due to the viscoelastic nature, lesser sphericity, and slightly higher average diameter of tissue spheroids compared to more rigid agarose beads. We demonstrated the ability to bioprint spheroids precisely (~11% with respect to the average spheroid size) onto alginate through patterning of various shapes (fig. S6, A1 to A3), initials of Penn State University (PSU) (Fig. 3A3), and a matrix of spheroids of various shapes and dimensions (including irregular shapes) in the diameter range of 80 to 200 m (made of HUVEC, 3T3, and 4T1 cells) (Fig. 3A4). Moreover, we also demonstrated the bioprinting of green fluorescent protein (GFP)labeled MSC spheroids in the shape of triangle and ring into other gels, including type I collagen (COL I) and gelatin methacryloyl (GelMA), as shown in Fig. 3 (A5 and A6, respectively). To highlight the unprecedented precision of AAB, we bioprinted eight different-sized MSC spheroids on top of each other without any gel support (Fig. 3, A7 and A8) and also fabricated a hollow bridge shape with MSC spheroids using the same approach (fig. S6, B1 and B2). Moreover, we demonstrated another example of a hollow bridge configuration, where the middle spheroid in the second layer was supported by the underlying gel without any spheroid support underneath (fig. S7). This was accomplished by bioprinting the gel and spheroids alternatingly at each layer, which necessitated the precise control of gel thickness; however, the gel thickness was dependent on several factors, such as the droplet resolution, cross-linking time, and spreading behavior of droplets, which were not trivial to control at the same time using microvalve bioprinting. To overcome this fundamental limit, self-healing Bingham plastic support gels (35) could be used to freely move the spheroids inside gels, which may bring more flexibility in self-assembly of highly complex geometries including hollowed-out structures.

In addition, self-assembly of spheroids was investigated at predetermined time points. As shown in Fig. 3B1, 3T3 spheroids were bioprinted into fibrin with close proximity and then cultured for 72 hours. The two individual 3T3 spheroids bioprinted next to each other gradually fused to form a larger spheroid to minimize their surface energy, as described in a previous study (21). The normalized contact length and intersphere angle increased by 50% over the first 24 hours of culture (Fig. 3B2). To evaluate the role of bioprinting on the viability of spheroids, two cases were evaluated. In case 1, the viability of spheroids was determined immediately after lifting spheroids and depositing them into another cell medium reservoir. In case 2, the viability of spheroids was evaluated after bioprinting them into the gel substrate. Spheroids, which were not subjected to the bioprinting process, were used as a positive control group. The viability of spheroids in the positive control group and case 1 were determined to be ~94 and ~88%, respectively (Fig. 3B3). In case 2, cell viability was measured to be ~82%. The decrease in cell viability could be due to the aspiration force or dehydration during the rapid transfer or cell damage when spheroids were submerged into the hydrogel substrate; however, viability levels over 80% for bioprinting of spheroids could still be considered moderate, as viability of fabricated cells aggregates even without bioprinting could be in that range (36).

In addition to bioprinting of spheroids, the AAB system also enabled the bioprinting of other biologics. For example, we demonstrated the bioprinting of electrocytes isolated from electrogenic organs of electric fish. Because of the asymmetric functionality of their anatomy and their spatial arrangements in series configuration in an electric organ (similar to batteries connected in series; Fig. 3C1), electrocytes produce a considerable amount of electricity (37). In this study, five electrocytes (~400 m in diameter; Fig. 3C2) from weakly electric fish (Brachyhypopomus gauderio) were obtained according to the animal protocol (Institutional Animal Care and Use Committee #47870) and patterned next to each other onto agarose using AAB with a back pressure of ~20 to 25 mmHg (Fig. 3C3). To the best of our knowledge, this is the first bioprinting of electrocytes, which can be used in biofabrication of biological batteries for various applicationssuch as pacemakers, cochlear implants, and brain chipsin the future (38). In addition, these cells could be arranged in the form of biological circuits for biocomputing or as bioelectric interfaces for use in treating limb loss, musculoskeletal disorders, and body augmentation as well as cyborg organs interfacing electrical devices and biology (39). The presented approach showed the possibilities of bioprinting not only spheroids or single cells but also irregularly shaped aggregates. For example, Fig. 3D shows vertical bioprinting of tissue strands [described in our previous work (5)] into the space between pins, which can be used for fabrication of scalable tissues of muscle, fat, cartilage, nerves, blood vessels, etc.

To demonstrate other unique capabilities of the presented approach, heterogeneous tissue complexes were 3D bioprinted using spheroids of different sizes and types including tdTomato+ HUVEC spheroids (~155 m) and GFP+ MSC spheroids (~390 m) (Fig. 4, A and B). Spheroids were arranged into a pyramid structure to clearly visualize the heterogeneous architecture using confocal imaging, where alginate was used as a sacrificial gel and the first, second, and third layers constituted of HUVEC, MSC, and HUVEC spheroids, respectively. This example clearly demonstrates the unique deposition ability of the AAB technique, as the last HUVEC spheroid was accurately placed into the small and confined space between three adjacent MSC spheroids (~2.5-fold of HUVEC spheroid size) on the third layer. Alginate preserved the structural integrity of the bioprinted pattern during the initial stage of culture. Moreover, it enabled the bioprinted entities to stick in their precise locations, which were defined before the bioprinting process. To print the sacrificial alginate, we used a unipolar wave pulse with a dwell voltage (amplitude) of 5 V and a dwell time (valve opening duration) of 1000 s for actuating the microvalve dispenser, where a positive back pressure of ~103 kPa was used for driving the flow of sodium alginate inside the tubing. The reader is referred to fig. S8A for the optimization of dwell time and droplet volume. Afterward, sodium alginate layer was partially cross-linked with aerosol form of CaCl2 for 30 s, and spheroids were bioprinted as explained in Fig. 1. Patterned spheroids were partially fused and maintained their initial position after 48 hours of incubation. After 2 days of culture, alginate was gently dissolved [using an alginate lyase as explained in our earlier work (5)], and the bioprinted constructs maintained their integrity even after the sacrificial alginate was removed. As evident from the confocal images (Fig. 4, C1 to C6), spheroids were well connected in 3D. A similar arrangement was also presented with MSC spheroids of three different dimensions, where spheroids got larger as more layers were laid down (fig. S9, A to C). We also demonstrated a more complex pattern such as a 3D diamond pattern with MSC/HUVEC spheroids (Fig. 4, D to G). To the best of our knowledge, this is the first bioprinting of tissue spheroids with such a positional accuracy, ~15% with respect to the spheroid size, yielding highly intricate geometries and heterogeneous structures in 3D.

(A) A schematic illustration of 3D bioprinting of a heterogeneous pyramid construct using different sizes and types of spheroids. (B) A photograph of the bioprinted three-layer heterogeneous pyramid. (C1 to C6) 3D reconstruction of confocal images of bioprinted pyramid of tdTomato+ HUVEC spheroids (first and third layers) and GFP+ MSC spheroids (second layer). (D) A schematic illustration of a diamond construct made of MSC/HUVEC spheroids. (E) A photograph showing 3D bioprinted diamond from the side camera. (F) Fluorescent images of the bioprinted diamond, which were stained with DAPI, CD31, and F-actin. (G1 to G4) Confocal images of the diamond construct (bottom view) (note that 1-day cultured HUVEC and 2-day cultured MSCs were used in these experiments). Photo credit: Bugra Ayan, Penn State University.

In this study, we have demonstrated the utilization of AAB in both scaffold-based and scaffold-free configurations and presented two unique applications demonstrating its potential in the development of physiologically relevant culture environments for studying angiogenic sprouting and fabrication of osteogenic tissues for tissue engineering purposes. To demonstrate the application of AAB in scaffold-based fabrication of physiologically relevant culture environments, we bioprinted HUVEC spheroids into fibrin hydrogel with a predetermined distance apart and studied their collective angiogenic sprouting behavior. This importantly signifies that angiogenic sprouting behavior can be tailored by varying the proximity of spheroids, hence directly affecting cell-cell signaling. To study the effect of distance on angiogenic sprouting, we deposited droplets of fibrinogen (6 mg/ml) [loaded with HDFs (0.5 million/ml)] and thrombin (2.4 U/ml) layer by layer (in 1:1 volume) to fabricate fibrin constructs, as such concentration ranges support angiogenesis of HUVECs (40). A positive back pressure of ~103 kPa was used for driving the flow of fibrinogen and thrombin solutions inside the tubing and dispenser. To dispense a ratio of 1:1 of fibrinogen and thrombin, we used dwell times of 500 and 700 s for obtaining the same droplet volumes at the microvalve dispensers, respectively. The reader is referred to fig. S8 (B and C) for the optimization of droplet generation for fibrinogen and thrombin solutions. HUVEC spheroids were then bioprinted into fibrin before complete gelation. The distances between the spheroids were maintained at 400 10, 800 13, and 3000 16 m (isolated spheroids used as a control group) (Fig. 5A1). Bioprinted constructs were cultured for a period of 7 days. Angiogenic sprouting from HUVEC spheroids at days 2, 5, and 7 was quantified using AngioTool software (41). Spheroids with close proximity to each other were seen to have higher total vessel length as compared to isolated spheroids right from the initial stages of culture. The combined length of sprouts was 13 and 11 mm for 400 and 800 m, respectively, as compared to the control group, which were around 6 mm (Fig. 5A2). A similar trend was also observed for the total vessel area. There was approximately 3- and 2.5-fold increase in the vessel area for 400- and 800-m distances on day 7 as compared to the control group, respectively. Although the total number of branching points or junctions was not significantly different among different groups at day 2, the sprouts branched more when spheroids were bioprinted close to each other, as compared to the control group (fig. S10). On day 7, the number of junctions for 400- and 800-m distances was 2.1- and 1.7-fold greater than those for the control group, respectively. The mean lacunarity refers to the amount of free space around the sprouted capillaries, and an increase in angiogenic sprouting results in a decrease in lacunarity (42). This yields a measurement of the sprouting activity, and the mean lacunarity was the highest for the control group compared to other two groups.

(A1) Epifluorescent images of bioprinted tdTomato+ HUVEC spheroids with varying distances (400 to 3000 m) apart on day 7. (A2) Graphical representation of various sprouting propertiesnamely, total vessels length, total number of junctions formed, vessel area, and mean lacunarityobtained at day 7 for bioprinted HUVEC spheroids (n = 3; ***P < 0.001). (B1) Epifluorescent images of bioprinted GFP+ and tdTomato+ HUVEC spheroids with varying distances (400 to 3000 m) apart on day 7 along with higher-magnification confocal images of the interface region in XY and YZ planes showing capillaries formed by both GFP+ and tdTomato+ HUVECs (indicated by white arrows). (B2) Directionality analysis demonstrating the direction and percentage of normalized number of sprouts on day 7 (n = 3; *P < 0.05, **P < 0.01, and ***P < 0.001) [note that green and red bars demonstrate the angles of interest (AOIs), which are [60, 60] for GFP+ HUVECs and [120, 240] for tdTomato+ HUVECs]. (B3) Confocal images at the interface of two spheroids showing capillaries formed by both GFP+ and tdTomato+ HUVECs. (C1) A schematic illustration of the directionality of sprouts from a HUVEC spheroid toward a spheroid of GFP+ HDF and MSC cocultures. (C2) The directionality analysis for different mixing ratios of HDF:MSC, including HDF (control), 2:1, 1:1, and MSC (control), on day 7 (n = 3; ***P < 0.001 shows significance between AOI and Other for each group, and #P < 0.05, ##P < 0.01, and ###P < 0.001 show significance among AOIs of different groups) [note that N/A represents the none applicability of the directionality analysis, as no sprouts were observed in the MSC-only group; no directionality analysis was performed for the 3000-m distance, as sprouting was not observed and HUVECs exhibited spreading only; 1-day cultured HUVECs were used in all experiments; and the critical lifting pressure for coculture HDF/MSC spheroids (2:1 and 1:1 ratio) was determined to be 28.7 and 29.1 mmHg, respectively, through interpolation of the critical lifting pressure values for HDF and MSC spheroids presented in Fig. 2F].

In our study, HUVEC spheroids bioprinted with 400- and 800-m distances apart were considered to be in close proximity. On the other hand, HUVEC spheroids bioprinted with 3000-m distance apart were far enough to be considered isolated spheroids (control group). Proximity between cells triggers a cascade of events that, in turn, influences cellular behavior, and to study these events, it is important to control localization under in vitro conditions (43). The presented approach offers the capability of controlling the distance between spheroids and thus aims to better understand this signaling between spheroids. Sprouting properties such as vessel length, junction formation, and vessel area were all enhanced when spheroids were close to each other as compared to when they were placed far apart. The presence of HDFs in the matrix surrounding HUVEC spheroids also reinforced the formation of stable sprouts. It is known that fibroblasts act as supporting cells and surround the capillary-like structures, which gradually develop by self-organization of endothelial cells and simultaneous degradation of fibrin matrix (44). Moreover, fibroblasts secrete various soluble growth factors such as vascular endothelial growth factor (VEGF), angiopoietin-1, platelet-derived growth factor (PDGF) (45, 46), which enhance and modulate the growth of angiogenic sprouts (47). Bioprinted HUVEC spheroids close to each other probably up-regulated the combined secretion of these growth factors in the vicinity of the spheroids, which led to enhanced sprouting properties. The sprouts from closely placed spheroids extended toward each other and gradually fused to form a capillary network.

To better understand whether the positioning of spheroids affected their collective angiogenic sprouting behavior, GFP+ and tdTomato+ HUVEC spheroids were bioprinted into fibrin gel (with same properties as discussed before) at 400-, 800-, and 3000-m distances apart, and cultured over a period of 7 days (Fig. 5B1). A directionality analysis was then performed to investigate the role of the presence of a nearby spheroid on the directionality of angiogenic sprouting. Thus, the angle made by each sprout with the horizontal line (0/180) was measured for all spheroids. The angle spans [60, 60] for GFP+ HUVEC and [120, 240] for tdTomato+ HUVEC spheroids, where sprouts primarily grew toward the other spheroid, were considered the angles of interest (AOIs), as highlighted in Fig. 5B2. On day 7, the percentage of normalized number of sprouts (where the total number of sprouts were normalized with respect to the total angle span) in AOI was significantly higher than that of the other angle span (indicated with Other) for 400- and 800-m distances, indicating a dominating directionality of angiogenic sprouting toward the other spheroid. The percentage of normalized number of sprouts for AOI was ~63 and 65 for GFP+ and ~59 and 61 for tdTomato+ HUVEC spheroids, for 400- and 800-m distances, respectively. Although the directionality of sprouts for all cases may not be very apparent on day 2 (initial stages of culture; fig. S11, A1 and A2), on day 5 (later stages of culture; figs. S11, B1 and B2, and S12), mature sprouts were more concentrated within AOI. In addition, for sprouts formed in directions falling outside the AOI (depicted in gray), we observed more random growth of sprouts over time. The sprouts for spheroids that were 3000 m apart were random and did not indicate any directionality from days 2 to 7 (Fig. 5B2, bottom, and fig. S12C). For 400-m distance, it was also observed that the GFP+ HUVECs suppressed the sprouting ability of tdTomato+ HUVECs day 2 onward, where higher numbers of GFP+ HUVEC sprouts were observed at the interface [see fluorescent images in fig. S11 (A1 to B1)]. Domination of GFP+ HUVEC sprouts could be due to the fact that GFP+ HUVECs in this experiment were from an earlier passage (passages 2 to 5) with respect to tdTomato+ HUVECs (passages 5 to 7) (48). In addition, we investigated whether sprouts from both spheroids interacted and thus performed confocal imaging at the interface close to the tdTomato+ HUVEC spheroid (Fig. 5B3). We noticed that some capillaries were formed by both GFP+ and tdTomato+ HUVECs, as shown by arrows, which could be due to vascular anastomosis or the migration of GFP+ HUVECs to the other side of the interface and their contribution to the capillary formation by tdTomato+ HUVECs spheroids. Overall, the results showed that HUVEC spheroids bioprinted close to each other influence each others sprouting behavior, which led to a well-organized network formation between the sprouting bodies.

In the abovementioned experiments, we investigated the role of multiple HUVEC spheroids on their collective sprouting behavior, where HDFs were bioprinted as a single-cell suspension in fibrin, had direct contact with HUVEC spheroids, acted as supporting cells, and aided in forming stable sprouts. To explore whether HDFs could still influence the sprouting behavior of HUVECs even when they were not in direct contact with HUVECs, we reconfigured the design of the experiments presented above. In this regard, a HUVEC spheroid was bioprinted along with a coculture spheroid of GFP+ HDFs and MSCs (with controlled distance apart), which enabled us to change the concentration of HDFs and study the differential effect of coculture composition on HUVEC sprouting. GFP+ HDF/MSC spheroids, in 2:1 and 1:1 ratios, were bioprinted next to tdTomato+ HUVEC spheroids in fibrin with the same properties as discussed before (Fig. 5C1). HDF and MSC (homocellular) spheroids were used as control groups. The distance between these spheroids were varied from 400 to 3000 m, as performed before. For the cases of 400 and 800 m, HUVECs were attracted toward spheroids of HDF, 2:1, and 1:1 groups and were seen arranging into sprouts over a period of 7 days (figs. S13 and S14). The directionality analysis on day 7 revealed that the percentage of normalized number of sprouts within AOI for HDF, 2:1, and 1:1 groups for 400-m distance were ~82, 80, and 67, respectively (Fig. 5C2). For 800-m distance, a similar directionality was observed, where the percentage of normalized number of sprouts within AOI for HDF, 2:1, and 1:1 groups were determined to be ~84, 81, and 76, respectively. On the other hand, spheroids in the MSC group (Fig. 5C2) or spheroids located apart with a 3000-m distance (fig. S15) did not induce sprouting day 2 onward; rather, HUVECs exhibited spreading behavior. On day 2, for the case of 800 m, although the tip cells from HUVEC spheroids were not observed to have any physical contact with HDFs at the interface of both spheroids, sprout-like structures were already formed (fig. S16A). These structures were directed toward HDFs, as the relevant directionality analysis revealed a significant difference between AOI and Other for HDF, 2:1, and 1:1 groups (fig. S16B). This could be due to the fact that HDFs communicated with HUVECs via paracrine signaling by the possible diffusion of secreted growth factors, such as VEGF, angiopoietin-1, and PDGF, as discussed before (45, 46). By day 5 when HDFs substantially migrated out of the coculture spheroids, HDFs interacted with HUVECs outside AOI, particularly within angle spans [60, 120] and [240, 300], and induced sprout formation in those directions (fig. S17). Overall, more stable sprouts were observed in HDF-involved spheroids, and the results revealed that better directionality of angiogenic sprouting could be attained when the density of HDFs increased in the coculture spheroid. The presence of MSCs, on the other hand, had a negative impact on angiogenic sprouting of HUVECs, which could be due to the inhibitory effect of MSCs on the angiogenic potential of HUVECs via cell-cell contact through modulation of the VE-cadherin/-catenin signaling pathway as described before (49, 50).

To demonstrate another major application of AAB, osteogenic tissues were bioprinted using MSC/HUVEC spheroids as building blocks in a scaffold-free configuration. After fabrication, two different osteogenic differentiation culture strategies were used, both with equal total exposure time of osteogenic induction. This was performed to decode the role of midterm osteogenic induction of stem cellbased spheroids (before bioprinting) on the mineralization and assembly behavior of generated tissues (fig. S18). In strategy no. 1, 2-day cultured spheroids were bioprinted into a triangle pattern (Fig. 6A). After bioprinting, triangle-shaped tissue complexes maintained their integrity and transformed into a more compact and dense structure after 3 days in proliferation culture conditions [growth media (GM)]. Spheroids in this triangle-shaped pattern self-assembled over time, where cells exhibited a viability of 83% after 5 days of incubation, and the assembled structure was further cultured for a period of 12 days in osteogenic media (OM) (Fig. 6, B and C). In general, MSCs cultured in OM express high levels of osteogenic markers (51). To confirm the osteogenic differentiation of bioprinted tissues, the early osteogenic differentiation marker, Runt-related transcription factor 2 (RUNX2), was used for immunohistochemical staining. At the same time, the presence of HUVECs was confirmed by CD31 staining. As shown in Fig. 6D, bioprinted tissues expressed osteogenic- and endotheliogenic-specific markers, as indicated by positive staining of RUNX2 and CD31. Calcium deposition of osteogenically differentiated tissues was also confirmed by Alizarin red staining. As shown in Fig. 6E, substantial calcium deposition was observed after 12 days of osteogenic induction, particularly at the core of the assembled tissue. As substantial contraction was observed after the fusion of MSC/HUVEC spheroids and the originally bioprinted triangular shape was not maintained (Fig. 6C) as well as the mineralization was not uniform throughout the tissue boundary (Fig. 6E), we also followed an alternative strategy (strategy no. 2) to preserve the bioprinted shape of osteogenic tissues (fig. S18). In this regard, MSC/HUVEC spheroids were first maintained in GM for 5 days, followed by inducting them with OM for 10 days. Next, spheroids were bioprinted and then kept for another 2 days in OM. In both strategies, the total exposure time to GM and OM was identical. At the end of culture (Fig. 6F), bioprinted tissues using strategy no. 2 exhibited strong expression of RUNX2 with more uniform distribution of mineralization demonstrated by Alizarin red staining, although RUNX2 staining was stronger in cores of spheroids (Fig. 6, G to J). Intensity analysis of RUNX2 revealed that RUNX2 staining was the strongest in the core of tissues or at the interface between spheroids and the weakest on the surface of the assembled tissues (Fig. 6, K and L, and fig. S19A), while CD31 staining was uniformly distributed throughout the histological sections of bioprinted tissues under both strategies (fig. S19B). A previous study demonstrated that HUVEC-mediated paracrine factors, including VEGF and the inflammatory mediator prostaglandin E2 (PGE2), promoted the osteogenesis of periodontal ligament stem cells under hypoxic conditions regulated by mitogen-activated protein kinase (MAPK) kinase/extracellular signalregulated kinase and p38 MAPK pathways (52). As the core of tissues and interface between spheroids were more hypoxic compared to the surface of assembled tissues, the osteogenic differentiation of MSCs could be further advanced in those regions by PGE2 and VEGF paracrine factors secreted by HUVECs. In addition, limited shape change was observed. This could be due to the fact that MSCs in MSC/HUVEC spheroids, cultured in OM for 10 days before bioprinting, were in the osteogenic differentiation pathway and their proliferation capability was diminished as reported in a previous study (53), and hence, limited contraction was observed in spheroids. Another advantage of strategy no. 2 is that the culture duration after bioprinting was reduced, where 2-day cultured tissues after bioprinting were still structurally stable. We also analyzed the expression levels of bone sialoprotein (BSP), type I collagen (COL1), alkaline phosphatase (ALP), RUNX2, and CDH2 (N-cadherin) genes for both strategies, and bioprinted tissues cultured in GM (labeled as 3D bioprinted tissue cultured with GM) and MSCs differentiated in 2D for 12 days (labeled as 2D MSCs cultured with OM) were used as control groups. As shown in Fig. 6M, expression levels of BSP, COL1, ALP, and RUNX2 genes for both strategies were similar to each other and significantly higher than those of control groups. In addition, the expression level of CDH2 gene (encoding N-cadherin protein) for all 3D bioprinted tissue groups were similar to each other and higher than that of the 2D control group. Overall, by changing the osteogenic induction window under the same total osteogenic induction duration, the shape of bioprinted tissues and uniformity of mineralization could be controlled, although no differences were detected in the expression levels of osteogenic genes.

Strategy no. 1: (A) Triangle-shaped tissue complexes were bioprinted using MSC/HUVEC spheroids and cultured for 3 days in GM and 12 days in OM. (B) Time-lapse images showing fusion of GFP+ spheroids up to day 15 (D15) after bioprinting. (C) An optical image showing the assembled tissue at day 15 after bioprinting. (D) Immunofluorescence staining (DAPI, CD31, F-actin, RUNX2, and DAPI + RUNX2) and (E) Alizarin red staining of the sectioned tissue. Strategy no. 2: (F) The final shape of the bioprinted tissue of osteogenic spheroids (cultured for 10 days in OM before bioprinting and 2 days in OM after bioprinting). Immunofluorescent images of (G) the bioprinted tissue and (H) confocal images of its histological sections stained for DAPI, CD31, and F-actin and (I) RUNX2 and DAPI + RUNX2. (J) Alizarin red staining of the tissue section. (K) Quantification of normalized RUNX2 intensity at different regions including the surface of assembled tissue, spheroid-spheroid interface, and core of spheroids (n = 50; **P < 0.01 and ***P < 0.001). (L) A representative heat map figure showing RUNX2/DAPI distribution in the surface of assembled tissue, spheroid-spheroid interface, and core of spheroids for strategy nos. 1 and 2. (M) BSP, COL1, ALP, RUNX2, and CDH2 gene expressions of 2D MSCs cultured in OM (control), 3D bioprinted tissues cultured in GM (control), and 3D bioprinted tissues cultured using strategy nos. 1 and 2 (n = 5; **P < 0.01 and ***P < 0.001).

Sodium alginate solution was made by dissolving 1% (w/v) sodium alginate (Sigma-Aldrich, UK) in deionized (DI) water. Calcium chloride solution was prepared by dissolving 4% (w/v) calcium chloride (CaCl2) (Sigma-Aldrich, St. Louis, MO) in DI water. For constructing fibrin scaffolds, fibrinogen (6 mg/ml) (Sigma-Aldrich) and thrombin (2.4 U/ml) (Sigma-Aldrich) were prepared for microvalve bioprinting. For bioprinting of electrocytes, agarose (A20070-100, Research Products International, IL) was dissolved in DI water to obtain a solution of 1% (w/v) agarose at 90C. COL I was extracted from rat tails according to a published protocol (54). GelMA was synthesized according to an established protocol (55). Detailed preparation methods of both materials can be found in the Supplementary Materials.

The mouse fibroblast cell line, 3T3, and the mouse mammary carcinoma line, 4T1, were obtained from the laboratory of A. Mastro, PSU (State College, PA). Culture media for 3T3 fibroblasts composed of Dulbeccos modified Eagles medium (DMEM) (Corning, Manassas, VA), supplemented with 10% fetal bovine serum (FBS) (Life Technologies, Grand Island, NY) and 1% penicillin-streptomycin (Corning). 3T3 cells were used at passages 22 through 27. 4T1 cells were grown in RPMI (Corning), 10% FBS, 1% penicillin-streptomycin. Passages 7 through 12 were used for 4T1 cells. HUVECs were purchased from Lonza and cultured in MCDB 131 base media (Corning) supplemented with 10% FBS (Corning), 1% glutamine (Gibco, Life Technologies), 0.5% bovine brain extract (Lonza, Walkersville, MD), heparin (10 U/ml) (Sigma-Aldrich), endothelial cell growth supplement (3 mg/ml) (Sigma-Aldrich), and 1% penicillin-streptomycin (Corning). HUVECs were used at passages 3 through 8. HUVECs were also transduced in house with EF1 tdTomato lentivector (Vectalys, Toulouse, France) to ease cell visualization for fluorescence microscopy according to the manufacturers instructions. GFP+ HUVECs were purchased from Angio-Proteomie (cAP-0001GFP; Boston, MA) and were used at passages 2 through 5. HDFs, obtained from N. Zavazavas laboratory at The University of Iowa (Iowa City, IA), were cultured in DMEM supplemented with 10% FBS (Corning), 1% glutamine (Gibco), 1% sodium pyruvate (Gibco), and 1% penicillin-streptomycin (Corning). HDFs were used at passages 7 through 12. GFP+ HDFs were purchased from Angio-Proteomie (cAP-0008-adGFP; Boston, MA) and were used at passages 2 through 6. MSCs were obtained from Lonza (Walkersville, MD) and RoosterBio (Frederick, MD) and cultured in SU-005 RoosterBasal-MSC (RoosterBio). Passages 4 through 8 were used for MSCs. GFP+ MSCs were purchased from Cyagen, cultured in SU-005 RoosterBasal-MSC (RoosterBio), and used at passages 2 through 6.

All cells were maintained at 37C in a 5% CO2 humidified atmosphere. Cell culture medium was changed every 2 to 3 days. Subconfluent cultures were detached from the flasks using a 0.25% trypsin0.1% EDTA solution (Life Technologies) and split to maintain cell growth.

Adherent cultures of each cell type were detached from the culture vessels with 0.25% trypsin-EDTA (Life Technologies, Grand Island, NY) solution. Trypsin was neutralized with appropriate GM, and cells were counted by a hemocytometer. Each cell type was then diluted to a concentration of 2500, 5000, and 10,000 cells in 200 l of appropriate GM. The cell suspension (200 l) was then pipetted into a single well of a U-bottom 96-well microplate with a cell-repellent surface (Greiner Bio-One, Monroe, NC). For fabrication of MSC/HUVEC spheroids made of 50,000 cells, MSCs and HUVECs were combined at a ratio of 92:8. The microplates were then incubated at 37C in a 5% CO2 humidified atmosphere. Spheroid formation was monitored daily on an EVOS FL cell imaging system (Life Technologies). For fabrication of GFP+ HDF/MSC coculture spheroids, 5000 cells were used in total, and GFP+ HDFs and MSCs were cocultured in ratios of 2:1 and 1:1 for 1 day. During the fabrication and culture of HUVECs spheroids, EGM-2MV medium (Lonza) was used.

Field-emission SEM (FEI Nova NanoSEM 630) was used to investigate the surface topography of fabricated spheroids and dissociated electrocytes (see the Supplementary Materials for dissociation of electrocytes). Spheroids were harvested after 2 days of culture. Spheroids and electrocytes were fixed in 4% paraformaldehyde (Sigma-Aldrich) overnight; samples were then carefully washed in phosphate-buffered saline (PBS) and dehydrated using graded ethanol solutions (25 to 100%). To ensure complete removal of water, samples were further dried in a critical point dryer (CPD300, Leica EM, Wetzlar, Germany). On complete dehydration, they were sputter-coated with iridium (Leica) and imaged at an accelerating voltage of 3 to 5 keV.

A total of 1000 2-day cultured spheroids of each type (with ~2500 cells per spheroid) were collected and homogenized in 100 l of distilled water. Then, 100 l of cell homogenate was transferred to a 2-ml pressure-tight vial, added with 100 l of 12 M hydrochloric acid, and hydrolyzed at 120C for 3 hours. The total collagen amount per spheroid type was quantified using a hydroxyproline colorimetric assay kit (BioVision Inc., CA) according to the manufacturers instructions. Collagen amount expressed by each spheroid type was determined using a PowerWave X-340 spectrophotometer (BioTek, Winooski, VT) at 560 nm, and the results were normalized to the collagen amount expressed by HUVEC spheroids at day 2. Experiments were repeated four times.

Bioprinting of spheroids was used with a MakerBot Replicator 1 3D printer (MakerBot, NY). The extrusion head was removed, and a holder for a pipette and two microvalve heads was 3D-printed using an Ultimaker 2 (Ultimaker) 3D printer. To control the 3D motion stage, a smoothie board (Uberclock, OR) was integrated. The reader is referred to the Supplementary Materials for the details on the construction of the AAB platform.

For fabrication of support constructs, microvalves (INKX0517500A, Lee Company, Bashville, TN) with 250-m nozzles (INZA3100914K, Lee Company) were integrated on the bioprinter head. To operate microvalves, a control board (IECX0501350A, Lee Company) was used, which was controlled using Arduino Uno (Arduino, Italy). To cross-link sodium alginate, a portable ultrasonic humidifier (CZHD20, Comfort Zone, China) was used to generate the aerosol form of CaCl2. Details about the bioprinter setupincluding the computer aided design model (fig. S1D), block diagram (fig. S1E), and computer interface (fig. S20)can be found in the Supplementary Materials.

Spheroids were collected into a petri dish from U-bottom 96-well plates, and then, their surface tension was measured using a micropipette aspiration technique, as explained in our recent work (8). Aspirated spheroids were monitored via an STC-MC33USB monochromatic camera (Sentech, Japan) equipped with 1-61448 and 1-61449 adaptor tubes (Navitar, Rochester, NY).

Spheroids fixed in 4% paraformaldehyde (Sigma-Aldrich) and rinsed in Dulbeccos PBS (DPBS) were stained with NucBlue ReadyProbes reagent (Life Technologies) to visualize the cell nuclei (Fig. 3, A3 and A4); actin cytoskeletal fibers were stained with ActinGreen 488 ReadyProbes reagent according to the manufacturers instructions. Imaging and 3D reconstruction of a heterogeneous pyramid structure (Fig. 4A) was performed using a Zeiss confocal microscope (LSM 880, Carl Zeiss AG, Oberkochen, Germany) using a 5 lens. The 3D reconstruction was obtained using Zen blue software (Carl Zeiss AG). A diamond structure (Fig. 4D) was first imaged through Zeiss Axiozoom (Carl Zeiss AG, Oberkochen, Germany) to capture the entire 3D structure and then further imaged on a confocal microscope (Olympus FV1000, Olympus America Inc., Center Valley, PA) to closely image the DAPI (4,6-diamidino-2-phenylindole), F-actin, and CD31 staining.

Spheroids were removed from the U-bottom well plates and subjected to the bioprinting process. Spheroids directly taken from plates were used as a positive control group. Viability was assessed using a LIVE/DEAD viability assay kit (Life Technologies, Grand Island, NY). Briefly, spheroids were washed twice in DPBS and then placed in the dye solution consisting of 1 M calcein acetoxymethyl and 1.6 M ethidium homodimer-1 in PBS. Live cells were able to take up and retain the calcein dye, resulting in bright green fluorescence of their cytoplasm. The ethidium homodimer could only enter dead cells where it binds to nucleic acids, producing a bright red fluorescence. Spheroids were imaged at 20-m z-stack step size on the Olympus FV1000 confocal microscope with a 60-m aperture setting resulting in approximately five to seven images per spheroid. Each image was then analyzed using ImageJ software (National Institutes of Health, USA).

Spheroids made of 3T3 cells were collected from U-bottom 96-well plates and then washed with DPBS twice. CellTracker Orange CMTMR [5-(and-6)-(((4-chloromethyl)benzoyl)amino) tetramethylrhodamine] (Thermo Fisher Scientific) and CellTracker Green CMFDA (5-chloromethylfluorescein diacetate) (Thermo Fisher Scientific, MA) were used according to the manufacturers instructions. Spheroids were bioprinted with close proximity, and then, images were captured at 0, 24, 48, and 72 hours using EVOS FL Cell Imaging System. The contact length and contact angle of two spheroids were measured every 4 hours up to 24 hours.

Images of sprouting GFP+ and tdTomato+ HUVEC spheroids at day 7 were taken on the Zeiss Axiozoom microscope at a magnification of 20 to capture the spheroids bioprinted at various distances. Images of sprouts at days 2 and 5 were taken using the EVOS FL Cell Imaging System. Sprouting behavior of spheroids was analyzed using AngioTool (41). To obtain the cross-sectional view of the capillary-like structures formed by GFP+ and tdTomato+ HUVEC spheroids at day 7, samples were imaged using a 40 oil lens on the Olympus FV1000 confocal microscope. For the study with GFP+ HDF/MSC coculture and HUVEC spheroids, samples were imaged on the EVOS microscope using fluorescent (4) channels at days 2 and 5. Images of those samples on day 7 were taken using fluorescent (16 and 40) and phase (16) channels by the Axiozoom microscope.

To induce osteogenic differentiation, MSC/HUVEC spheroids were bioprinted and cultured in two different strategies in a custom culture media made of 92% osteogenic differentiation media (Cell Applications Inc., San Diego, CA) and 8% HUVEC culture media. To confirm the morphology of bioprinted tissues undergoing osteogenic differentiation, tissues were sectioned and stained with RUNX2, CD31, F-actin, and DAPI. Cross-sectioned samples were washed three times with DPBS, fixed in 4% paraformaldehyde for 60 min, permeabilized in 0.2% Triton X-100 for 30 min, and blocked with 2.5% normal goat serum (NGS) for 60 min at room temperature. To visualize osteogenic and endothelial-specific genes, the samples were incubated with mouse anti-RUNX2 primary antibody (1:100 in 2.5% NGS) and rabbit anti-CD31 primary antibody (1:100 in 2.5% NGS) for 60 min; washed three times with DPBS; and incubated with goat anti-mouse Alexa Fluor 488 secondary antibody (Molecular Probes; 1:250 in 2.5% NGS) to label RUNX2, goat anti-rabbit Alexa Fluor 647 secondary antibody (Molecular Probes; 1:250 in 2.5% NGS) to label CD31, Alexa Fluor 568 phalloidin (Molecular Probes; 1:1000 in 2.5% NGS) to label filamentous actin, and DAPI (1:1000 in 2.5% NGS) to visualize cell nuclei for 60 min. The stained samples were washed three times with DPBS and imaged by an Olympus FV10i-LIV Confocal Laser Scanning Microscope (Olympus America Inc.) and analyzed using ImageJ software. In addition to sectioned samples, bioprinted tissues were also stained, as a whole-mount sample, as explained above, and fluorescent images were taken on the Zeiss Axiozoom microscope.

To confirm the calcium deposition, cross-sectioned samples were fixed in 4% paraformaldehyde, washed three times with distilled water, and incubated with 2% Alizarin red S stain solution for 30 min at room temperature. Stained samples were washed three times with distilled water and imaged using the EVOS microscope.

To quantify the intensity of RUNX2 and CD31, areas of interest on confocal images were selected using ImageJ. Fifty regions of interest were determined at the surface of the assembled tissue, spheroid-spheroid interface, and core of spheroids on fluorescent images. Each box was used for quantification of the fluorescence intensity. The intensity of RUNX2 and CD31 was normalized by the intensity of DAPI. A representative heat map was generated for the RUNX2/DAPI intensity.

Real-time polymerase chain reaction (PCR) was performed to quantify the gene expression levels of BSP, COL1, ALP, RUNX2, and CDH2. The primers of the measured mRNA genes were as follows: BSP (forward, AAC GAA GAA AGC GAA GCA GAA and reverse, TCT GCC TCT GTG CTG TTG), COL1 (forward, ATG ACT ATG AGT ATG GGG AAG CA and reverse, TGG GTC CCT CTG TTA CAC TTT), ALP (forward, AGC TGA ACA GGA ACA ACG TGA and reverse, CTT CAT GGT GCC CGT GGT C), RUNX2 (forward, GGT TAA TCT CCG CAG GTC ACT and reverse, CAC TGT GCT GAA GAG GCT GTT), CDH2 (forward, GAG CAG TGA GCC TGC AGA TTT T and reverse, TGC TCA GAA GAG AGT GGA AAG CT), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) (forward, ATG GGG AAG GTG AAG GTC G and reverse, GGG GTC ATT GAT GGC AAC AAT A). Real-time PCR was analyzed by using SsoFast EvaGreen Supermix (Bio-Rad, USA), and all results were normalized using GAPDH.

All data were presented as means SD and analyzed by Minitab 17.3 (Minitab Inc., State College, PA, USA) using one-way analysis of variance (ANOVA) to test for significance when comparing the data. Post hoc Tukeys multiple-comparison test was used to determine the individual differences among the groups. Differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001. For directionality analysis, t test was used to compare the results between AOI and Other (where differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001), and ANOVA (with Tukeys multiple comparison test) was used to compare the results among different groups (where differences were considered significant at #P < 0.05, ##P < 0.01, and ###P < 0.001). Repeated-measures ANOVA (with Tukeys multiple-comparison test) was conducted to compare the results among different days of same groups, and differences were considered significant at *P < 0.05, **P < 0.01, and ***P < 0.001.

See original here:
Aspiration-assisted bioprinting for precise positioning of biologics - Science Advances

Molecular Signature of Young-Onset Parkinson’s Disease Is… : Neurology Today – LWW Journals

Article In Brief

A unique molecular structureevident in induced pluripotent stem cells taken from people with young-onset Parkinson's diseasesuggests that the defects may be present throughout patients' lives, and that they could therefore be used as diagnostic markers.

Induced pluripotent stem cells (iPSCs) taken from patients with young-onset Parkinson's disease (YOPD) and grown into dopamine-producing neurons displayed a molecular signature that was corrected in vitro, as well as in the mice striatum, by a drug already approved by the US Food and Drug Administration (FDA), a study published in the January 27 online edition of Nature Medicine found.

Although the patients had no known genetic mutations associated with PD, the neurons grown from their iPSCs nonetheless displayed abnormally high levels of soluble alpha-synucleina classic phenotype of the disease, but one never before seen in iPSCs from patients whose disease developed later in life. Surprisingly, for reasons not yet understood, the cells also had high levels of phosphorylated protein kinase C-alpha (PKC).

In addition, the cells also had another well-known hallmark of PD: abnormally low levels of lysosomal membrane proteins, such as LAMP1. Because lysosomes break down excess proteins like alpha-synuclein, their reduced levels in PD have long been regarded as a key pathogenic mechanism.

When the study team tested agents known to activate lysosomal function, they found that a drug previously approved by the FDA as an ointment for treating precancerous lesions, PEP005, corrected all the observed abnormalities in vitro: it reduced alpha-synuclein and PKC levels while increasing LAMP1 abundance. It also decreased alpha-synuclein production when delivered to the mouse striatum.

Unexpectedly, however, PEP005 did not work by activating lysosomal function; rather, it caused another key protein-clearing cellular structure, the proteasome, to break down alpha-synuclein more readily.

The findings suggest that the defects seen in the iPSCs are present throughout patients' lives, and that they could therefore be used as diagnostic markers. Moreover, the drug PEP005 should be considered a potentially promising therapeutic candidate for YOPD and perhaps even for the 90 percent of PD patients in whom the disease develops after the age of 50, according to the study's senior author, Clive Svendsen, PhD, director of the Cedars-Sinai Board of Governors Regenerative Medicine Institute and professor of biomedical sciences and medicine at Cedars-Sinai.

These findings suggest that one day we may be able to detect and take early action to prevent this disease in at-risk individuals, said study coauthor Michele Tagliati, MD, FAAN, director of the movement disorders program and professor of neurology at Cedars-Sinai Medical Center.

But the study still raises questions regarding the biological mechanisms, and certainly does not warrant off-label prescribing of PEP005 at this time, said Marco Baptista, PhD, vice president of research programs at the Michael J. Fox Foundation, who was not involved with the study.

Repurposing PEP005 is a long way away, Dr. Baptista said. This is not something that neurologists should be thinking about prescribing or recommending to their patients.

Accumulation of alpha-synuclein has been seen in iPSC-derived dopaminergic cultures taken from patients with known genetic defects, but such defects account for only about 10 percent of the PD population. In those without known mutations, on the other hand, no defects in iPSC-derived dopamine-producing neurons have been seen. Until now, however, such studies had been conducted only in patients who had developed PD after age 50.

My idea was why to look in young-onset patients, said Dr. Svendsen.

The idea paid off more richly than he expected. We were shocked to find a very, very prominent phenotype, a buildup of alpha-synuclein, in the neurons of these patients who are genetically normal, Dr. Svendsen said. None of the controls had a buildup of synuclein, and all but one of the early PD patients had a twofold increase in it.

The signature is so consistent, he said, that it offers a natural model that can be interrogated to further understand its workings.

Because high levels of PKC were also seen, Dr. Svendsen said, We picked a bunch of drugs known to reduce PKC. We found one, PEP005, which is actually extracted from the milkweed plant, and it completely reduced synuclein levels almost to normal in dopaminergic neurons. And it also increased dopamine levels in those cells, so we got two for one.

After observing the effects of PEP005 in vitro, We put it into the mouse brain and found it reduced synuclein in vivo, Dr. Svendsen said. But we had to infuse it right into the brain. We're now trying to work out how to get it across the blood-brain barrier more efficiently.

To determine how PEP005 lowers cellular levels of alpha-synuclein, his group tested whether it was activating the lysosome, but found to their surprise that it did not do this until after the synuclein had already been degraded.

Then we asked whether it could be the proteosome, which also breaks down proteins but normally doesn't break down synuclein, Dr. Svendsen said. But when we applied PEP005, it did activate the proteasome. So we think that might be the mechanism.

Because the drug is currently applied externally, Dr. Svendsen said, the next step will be to see if it crosses the blood-brain barrier when applied to the skin of mice, and whether that results in a lowering of synuclein levels in dopaminergic neurons.

Justin Ichida, PhD, the Richard N. Merkin assistant professor of stem cell biology and regenerative medicine at the USC Keck School of Medicine, said the findings are quite important in the field. The potential diagnostic tools they made could be important in clinical care. And identifying a drug that may very effectively reverse the disease in neurons is a very important discovery.

He wondered, however, whether the increase in alpha-synuclein is truly specific to Parkinson's neurons or if it would also be seen in iPSC neurons from patients with Alzheimer's disease or amyotrophic lateral sclerosis.

I wonder if alpha-synuclein accumulating is a sign of PD in a dish or is a consequence of neurodegeneration or impaired protein degradation in general, Dr. Ichida said. That's a key question if you want to use this molecular signature as a diagnostic tool.

He also questioned if proteins other than alpha-synuclein, such as tau, would also be seen to accumulate in the iPSCs of YOPD patients.

If one of the protein-clearance mechanisms in the cell is working poorly, you would imagine that other things would also accumulate, Dr. Ichida said.

In response, Dr. Svendsen said that while some proteins other than alpha-synuclein were reported in the paper at increased levels, We did not look at tau specifically, but are in the process of looking right now. It could be that synuclein and some other proteins are somehow altered to evade them from being degraded by the lysosome, or that there is a general lysosomal problem.

Patrik Brundin, MD, PhD, director of the Center for Neurodegenerative Science and Jay Van Andel Endowed Chair at Van Andel Research Institute in Grand Rapids, MI, called the paper very interesting and thought-provoking. If these findings hold up, they could shift our understanding of young-onset PD. They imply that there is a strong genetic component that has not been picked up in prior genetic studies.

Dr. Brundin said he would like to see the results replicated in another lab using different sets of reagents. It is so intriguing and rather unexpected that one wonders if the observations really apply, as the study states, to 95 percent of all YOPD.

He also questioned whether all the young-onset PD patients are similar. Clearly the iPSCs studied here are not monogenetic PD, so they must be very diverse genetically and still all have the same alpha-synuclein change.

Dr. Brundin also asked why the abnormalities seen in YOPD neurons have not previously been seen in older cases of PD. Is there a specific cutoff regarding age-of-onset when these purposed genetic differences apply? he asked.

Dr. Svendsen responded: We don't know why the YO have this phenotype or exactly what the cut off is. We have, however, looked at one adult-onset case that did not show this phenotype. Also, one of our YO cases did not show this phenotype. Thus some patients even with early onset may not have it. We are currently testing many more cases from older-onset patients.

Dr. Brundin also wanted to know whether non-dopaminergic neurons have the same deficits described in the study.

We don't know which neurons specifically have the protein deficit as we cannot do single-cell proteomics, Dr. Svendsen answered. It could be a little in all cells or a lot in a small set. Immunocytochemistry is not quantitative but showed that it is more likely a general increase in synuclein and not specific to dopaminergic neurons.

While the findings in iPSCs suggest that the abnormal levels of alpha-synuclein must be present at birth, Dr. Brundin said, I do not know how to reconcile the present findings with genetic data.

The absence of previously described mutations in the YOPD patients means only that more work must be done to uncover the genetic underpinnings, Dr. Svendsen said.

We're just at the tip of the iceberg with understanding the genome, he said. It's such a bizarrely complex beast. Perhaps there are a thousand different proteins interacting to stop the synuclein from being degraded. In 10 years, we probably will be clever enough to see it. We know it must be there. Now the genome guys will go after it.

Dr. Baptista from the Michael J. Fox Foundation said he agreed with the view that there must be genetic alterations underpinning the defects seen in the iPSCs.

Just because we call something non-genetic could simply reflect the current ignorance of the field, he said. I think the discoveries are simply difficult to make.

He added that he wished that the main comparator in the study was not healthy controls, and that there were more older-onset iPSCs to compare against YOPD patients' samples.

Dr. Svendsen said it could be that the iPSCs from older-onset patients might yet be found with additional study to display abnormalities similar to those seen in YOPD.

Right now we only see it in young onset, he said. We may need to leave the cultures longer to see in the older-onset patients. We are doing those experiments now.

Drs. Tagliati and Svendsen disclosed that an intellectual patent is pending for diagnostic and drug screening for molecular signatures of early-onset Parkinson's disease. Dr. Ikeda is a co-founder of AcuraStem Inc. Dr. Brundin has received commercial support as a consultant from Renovo Neural, Inc., Lundbeck A/S, AbbVie, Fujifilm-Cellular Dynamics International, Axial Biotherapeutics, and Living Cell Technologies. He has also received commercial support for research from Lundbeck A/S and Roche and has ownership interests in Acousort AB and Axial Biotherapeutics. Dr. Baptista had no disclosures.

See the original post:
Molecular Signature of Young-Onset Parkinson's Disease Is... : Neurology Today - LWW Journals

How industry hopes to take on COVID-19 – Bioprocess Insider – BioProcess Insider

The biopharma space has stepped up its efforts to both prevent and treat the coronavirus (SARS-CoV-2) that is threatening to bring the world to its knees.

A month is a very long time when it comes to infectious diseases. The first cases and deaths from the novel coronavirus (COVID-19) led to a response to contain the virus, but the difficulties of containment and the nature of international travel means cases and deaths have become global.

The latest statistics place the number of cases at 95,483 and deaths at 3,286 across 84 countries, though by the time you are reading this the number is likely to have skyrocketed.

So as the world tilters on the edge of a pandemic, we take a look at how industry is responding. There is no specific treatment for the virus, nor a vaccine, but a proactive response is seeing the pharma industry throw everything in its arsenal at attempting to stymie this global threat.

First off, vaccines. As the World Health Organization (WHO) states it can take a number of years for a new vaccine to be developed, it has not stopped companies and academia stepping up their R&D efforts.

Both Sanofi and J&J have separately teamed up with the US Department of Health and Human Services (HHS) to expediate vaccine development.

Sanofi Pasteur aims to reverse engineer proteins isolated from the virus to produce DNA sequences, which will then be mass produced using Sanofi Pasteurs baculoviral expression system and formulated into a vaccine that elicits an immune response. Well that is the aim.

Johnson & Johnsons unit Janssen Pharmaceutical, meanwhile, is reviewing products in development for Middle East Respiratory Syndrome (MERS) or Severe Acute Respiratory Syndrome (SARS), to identify promising candidates for the novel coronavirus, and aims to upscale production and manufacturing capacities, leveraging its AdVac and PER.C6 technologies.

Another Big Vaccine company, GlaxoSmithKline, has teamed with Chinese biotech Clover Biopharmaceuticals to help develop a preclinical protein-based vaccine candidate. GSK will provide its pandemic adjuvant system for further evaluation of Clovers S-Trimer, a trimeric SARS-CoV-2 spike (S)-protein subunit vaccine candidate produced using a mammalian cell-culture based expression system.

Inovio Pharmaceuticals has also entered the race, and like GSK has teamed up with a Chinese company. Together with Beijing Advaccine Biotechnology and a grant of up to $9 million from the Coalition for Epidemic Preparedness Innovations (CEPI), Inovio hopes to bring its DNA vaccine candidate INO-4800 rapidly into clinical trials. VGXI a subsidiary of GeneOne Life Science has been selected to manufacture the DNA vaccine from its facilities in The Woodlands, Texas.

Thegenome sequence for 2019-nCoVwas published on January 10, 2020, a VGXI spokesperson recently toldBioprocess Insider. This DNA sequence information is used by Inovio and their collaborators at the Wistar Institute to design a synthetic DNA plasmid for manufacturing at VGXI. No viral particles or proteins are involved in the manufacturing process. When delivered as a vaccine, the DNA plasmid can elicit a protective immune response.

RNA vaccines are also being investigated. Moderna Therapeutics recently shipped the first batch of its investigational messenger RNA vaccine mRNA-1273 to the National Institute of Allergy and Infectious Diseases (NIAID) for use in a Phase I study. The vaccine is designed to train the immune system to recognize cells invaded by the coronavirus.

Moderna also received a grant from CEPI, as has CureVac, which is looking to use its mRNA vaccine platform to expedite a candidate into trials. CureVacs technology and mRNA platform are especially suitable to rapidly provide a response to a viral outbreak situation like this, said CureVac CTO Mariola Fotin-Mleczek. Currently, we are in the process of developing a vaccine that, after successful preclinical tests, could be tested rapidly in humans in a clinical study.

But industry could be pipped to the clinical trial post by academia, with Israels MIGAL Research Institute claiming to be sitting on a human vaccine against COVID-19 as a by-product of a vaccine it has developed against avian coronavirus Infectious Bronchitis Virus (IBV).

From research conducted at MIGAL, it has been found that the poultry coronavirus has high genetic similarity to the human COVID-19, and that it uses the same infection mechanism, a fact that increases the likelihood of achieving an effective human vaccine in a very short period of time, the Institute says.

According to MIGALs Biotechnology group leader Chen Katz, the vaccine is based on a new protein expression vector, which forms and secretes a chimeric soluble protein that delivers the viral antigen into mucosal tissues by self-activated endocytosis a cellular process in which substances are brought into a cell by surrounding the material with cell membrane, forming a vesicle containing the ingested material causing the body to form antibodies against the virus.

Other pharma companies are looking to treat coronavirus, rather than prevent.

Regeneron has teamed with the HHS to use its VelociSuite technologies to identify and validation and develop preclinical candidates and bring them to development, having followed a similar approach to advance its investigational Ebola treatment REGN-EB3.

The tech platform includes the VelocImmune mouse technology, a genetically modified strain in which genes encoding mouse immune system proteins have been replaced by their human equivalents.

The life-saving results seen with our investigational Ebola therapy last year underscore the potential impact of Regenerons rapid response platform for addressing emerging outbreaks, said George Yancopoulos, Regeneron CSO. Our unique suite of technologies expedites and improves the drug discovery and development process at every stage, positioning Regeneron to respond quickly and effectively to new pathogens.

Meanwhile this week, Takeda announced it is looking to a therapy to target COVID-19 based on polyclonal hyperimmune globulin (H-IG). The candidate, TAK-888, aims to concentrate pathogen-specific antibodies from plasma collected from recovered patients. Initially, due to a lack of current donors, the firm will produce the therapy in a segregated area within its manufacturing facility in Georgia.

The Japan-headquartered firm will also review its current pipeline for any other viable candidates to take on COVID-19.

Such an approach has aided Gilead Sciences efforts. The firm has begun two Phase III clinical studies of its antiviral candidate remdesivir, developed (though never approved) to treat Ebola virus. It has also shown promise against other infectious diseases including Marburg, MERS and SARS.

This is an experimental medicine that has only been used in a small number of patients with COVID-19 to date, so Gilead does not have an appropriately robust understanding of the effect of this drug to warrant broad use at this time, Gilead said.

With about 1,000 patients set to be tested with remdesivir, Gilead has turned to a stockpile manufactured in response to Ebola to address present coronavirus needs, and in anticipation of expanded use is manufacturing two formulations of remdesivir, in both liquid and freeze-dried forms, while upping capacity and production internally and externally.

According to San Marinos Bioscience Institute SpA, a regenerative medicine center and stem cell production facility, mesenchymal stem cells could potentially be treatment for the novel coronavirus by improving lung microenvironment, inhibiting immune system overactivation, promoting tissue repair, protecting lung alveoli epithelial cells, preventing pulmonary fibrosis, and improving lung function.

The company, citing the Chinese open repository for scientific researchers chinaXiv.org , says at least 14 trials are taking place in China using stem cells to treat coronavirus patients after positive animal testing showed stem cells might be able to repair the severe organ damage caused by the virus.

The firm even reports that a critically ill 65-year-old Chinese woman infected with SARS-CoV-2, whose conditions significantly improved after the infusion of mesenchymal stem cells.

If mesenchymal stem cells do prove to be the solution to the potential coronavirus crisis, Bioscience Institute alludes to the advantage that they are obtained from fat cells.

That means that everyone can utilize his/her cells, eliminating any contamination or rejection risk, said Giuseppe Mucci, CEO of Bioscience Institute.

But expanding them to the quantity needed for infusion, that corresponds to at least 1 million cells per kg of weight, takes 2 to 3 weeks. That is why it is useful to cryopreserve a personal reserve of mesenchymal stem cells, that would allow to access an early, more successful, treatment.

Read the original post:
How industry hopes to take on COVID-19 - Bioprocess Insider - BioProcess Insider

Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 – 2027 – Yahoo…

Cell Therapy Market Size, Share & Trends Analysis Report By Use-type (Research, Commercialized, Musculoskeletal Disorders), By Therapy Type (Autologous, Allogeneic), By Region, And Segment Forecasts, 2020 - 2027

New York, March 05, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 - 2027" - https://www.reportlinker.com/p05868803/?utm_source=GNW

The global cell therapy market size is expected to reach USD 8.8 billion by 2027 at a CAGR of 5.4% over the forecast period. Cellular therapies hold a great therapeutic promise across various clinical applications. This has resulted in substantial global investments in research and clinical translation. Moreover, rapid advances in stem cell research hold the potential to fulfill the unmet demand of pharmaceutical entities, biotech entities, and doctors in disease management. These factors have boosted revenue growth for the market.

Currently, there are a limited number of FDA-approved commercial stem and non-stem cell therapies in the market.Furthermore, LAVIV (Azficel-T), manufactured and commercialized by Fibrocell Technologies, witnessed revenue wind-down in the past years.

Key developers are making substantial investments in the adoption of advanced technologies to address the aforementioned challenges.

The introduction of proprietary cell lines is recognized as the primary means by which a single cell can be exploited for the production of a robust portfolio of candidates. Companies are leveraging new technologies not only for the expansion of their product portfolio but also for establishing out-licensing or co-development agreements with other entities to support their product development programs.

For instance, MaxCyte has more than 40 high-value cellular therapy partnership programs within immune-oncology, regenerative medicine, and gene editing, including fifteen clinical-stage programs. Increase in the number of collaborations between entities for product commercialization is anticipated to accelerate market revenue to a major extent in the coming years.

In Asia Pacific, the market is anticipated to witness significant growth over the forecast period.This is attributed to rising awareness cellular therapies among patients and healthcare entities in chronic disease management.

In addition, availability of therapeutic treatment at lower prices is also driving the regional market. Japan is likely to witness fast growth over the forecast period attributed to increasing research activities on regenerative medicine.

Further key findings from the report suggest: The clinical-use segment accounted for low revenue share due to stringent regulations and non- commercial viability of some products However, the expanding knowledge over the commercial potential of cellular therapies is anticipated to result in the commercialization of a large number of products in the coming years On the contrary, the research-use segment accounted for the largest revenue share in 2019 owing to increase in research activities to explore the potential of the therapy in substantially improving disease management Furthermore, an increase in funding to explore the potential of these therapies has contributed to the large share of the research segment Allogenic therapies dominated the revenue share in 2019 owing to relatively lower relapse rates and growth in stem cell banking activities This is due to the high price and a large number of companies involved in the development of allogenic therapies Moreover, several companies are preparing to shift their business towards allogeneic therapy product development, resulting in significant revenue growth in this segment Autologous therapies are estimated to grow at the fastest pace during the forecast period Lack of donors and low affordability of allogeneic therapies are two key factors contributing to the increase in adoption of autologous therapies Considering the growing share of the cell therapy market in the biopharma industry, the companies are striving to gain a competitive advantage Vericel Corporation, JCR Pharmaceuticals Co. Ltd., MEDIPOST, and Osiris Therapeutics, Inc. are some key players operating in the market These companies are engaged in the expansion of their product portfolio, either through product development or acquisition of other players operating in the space.Read the full report: https://www.reportlinker.com/p05868803/?utm_source=GNW

About ReportlinkerReportLinker is an award-winning market research solution. Reportlinker finds and organizes the latest industry data so you get all the market research you need - instantly, in one place.

__________________________

Story continues

Clare: clare@reportlinker.comUS: (339)-368-6001Intl: +1 339-368-6001

Original post:
Cell Therapy Market Size, Share & Trends Analysis Report By Use-type, By Therapy Type, By Region And Segment Forecasts, 2020 - 2027 - Yahoo...

With Over 280 Therapies Under Evaluation, the Stem Cell Therapy Market is Estimated to be Worth USD 8.5 Billion by 2030, Claims Roots Analysis – Yahoo…

The success of approved stem cell therapies has caused a surge in interest of biopharma developers in this field; many innovator companies are currently progressing proprietary leads across different phases of clinical development, with cautious optimism

LONDON, March 4, 2020 /PRNewswire/ -- Roots Analysishas announced the addition of "Global Stem Cells Market: Focus on Clinical Therapies, 20202030 (Based on Source (Allogeneic, Autologous); Origin (Adult, Embryonic); Type (Hematopoietic, Mesenchymal, Progenitor); Lineage (Amniotic Fluid, Adipose Tissue, Bone Marrow, Cardiosphere, Chondrocytes, Corneal Tissue, Cord Blood, Dental Pulp, Neural Tissue Placenta, Peripheral Blood, Stromal Cells); and Potency (Multipotent, Pluripotent))" report to its list of offerings.

There is a growing body of evidence supporting the vast applicability and superiority of treatment outcomes of stem cell therapies, compared to conventional treatment options. In fact, the unmet needs within this domain have spurred the establishment of many start-ups in recent years.

To order this 500+ page report, which features 185+ figures and 220+ tables, please visit this link

Key Market Insights

Over 280 stem cell therapies are under development, most of which are allogeneic products

More than 50% of the pipeline candidates are in the mid to late phase trials (phase II and above), and allogenic therapies (majority of which are derived from the bone marrow) make up 65% of the pipeline.

70% of pipeline candidates are based on mesenchymal stem cells

It is worth highlighting that the abovementioned therapies are designed to treat musculoskeletal (22%), neurological (21%) and cardiovascular (15%) disorders. On the other hand, hematopoietic stem cell-based products are mostly being evaluated for the treatment of oncological disorders, primarily hematological malignancies.

Close to 85% stem cell therapy developers are based in North America and Asia-Pacific regions

Within these regions, the US, China, South Korea and Japan, have emerged as key R&D hubs for stem cell therapies. It is worth noting that majority of the initiatives in this domain are driven by small / mid-sized companies

Over 1,500 grants were awarded for stem cell research, since 2015

More than 45% of the total amount was awarded under the R01 mechanism (which supports research projects). The NCI, NHLBI, NICHD, NIDDK, NIGMS and OD emerged as key organizations that have offered financial support for time periods exceeding 25 years as well.

Outsourcing has become indispensable to R&D and manufacturing activity in this domain

Presently, more than 80 industry / non-industry players, based in different regions across the globe, claim to provide contract development and manufacturing services to cater to the unmet needs of therapy developers. Examples include (in alphabetical order) Bio Elpida, Cell and Gene Therapy Catapult, Cell Tech Pharmed, GenCure, KBI Biopharma, Lonza, MEDINET, Nikon CeLL innovation, Roslin Cell Therapies, WuXi Advanced Therapies and YposKesi.

North America and Asia-Pacific markets are anticipated to capture over 80% share by 2030

The stem cell therapies market is anticipated to witness an annualized growth rate of over 30% during the next decade. Interestingly, the market in China / broader Asia-Pacific region is anticipated to grow at a relatively faster rate.

Story continues

To request a sample copy / brochure of this report, please visit this link

Key Questions Answered

The USD 8.5 billion (by 2030) financial opportunity within the stem cell therapies market has been analyzed across the following segments:

The report features inputs from eminent industry stakeholders, according to whom stem cell therapies are currently considered to be a promising alternatives for the treatment of a myriad of disease indications, with the potential to overcome challenges associated with conventional treatment options. The report includes detailed transcripts of discussions held with the following experts:

The research covers brief profiles of several companies (including those listed below); each profile features an overview of the company, financial information (if available), stem cell therapy portfolio and an informed future outlook.

For additional details, please visit

https://www.rootsanalysis.com/reports/view_document/stem-cells-market/296.htmlor email sales@rootsanalysis.com

You may also be interested in the following titles:

Contact:Gaurav Chaudhary+1(415)800-3415+44(122)391-1091Gaurav.Chaudhary@rootsanalysis.com

View original content:http://www.prnewswire.com/news-releases/with-over-280-therapies-under-evaluation-the-stem-cell-therapy-market-is-estimated-to-be-worth-usd-8-5-billion-by-2030--claims-roots-analysis-301016239.html

SOURCE Roots Analysis

Follow this link:
With Over 280 Therapies Under Evaluation, the Stem Cell Therapy Market is Estimated to be Worth USD 8.5 Billion by 2030, Claims Roots Analysis - Yahoo...

Stemina’s Human Stem Cell-Based Test Demonstrates Potential to Predict Which Drugs and Chemicals Cause Birth Defects – Business Wire

MADISON, Wis.--(BUSINESS WIRE)--Recently published results from an evaluation of 1,065 chemical and drug substances using the devTOX quickPredict (devTOXqP) screening platform developed by Stemina Biomarker Discovery, Inc. demonstrated the platforms ability to predict developmental toxicity in humans with high accuracy using a cell-based test. In a peer-reviewed article published in Toxicological Sciences, scientists from the U.S. Environmental Protection Agency (EPA) reported that Steminas devTOXqP test predicted the potential for developmental toxicity in a blinded set of chemicals and drugs from the agencys ToxCast program with an accuracy of 82%, where there was clear evidence of toxicity in humans or in animal studies. The agencys research suggests that devTOXqP is a useful tool for predicting developmental toxicants in humans and reducing the need for animal testing.

Through this EPA research, the devTOXqP test demonstrated its potential to detect developmental toxicity across a wide variety of chemicals and pharmaceutical compounds, said Elizabeth Donley, J.D., M.B.A., M.S., chief executive officer of Stemina. In addition to helping meet the EPAs goal of reducing the use of animals in testing chemicals, devTOXqP offers the only species-specific, commercially available platform for evaluating a drug or chemicals potential to cause birth defects in the developing human embryo. We believe this test fits well into global initiatives such as Tox21 and REACH that are seeking to reduce and refine the number of animals used for toxicity testing.

The potential of chemical substances to cause prenatal developmental toxicity is commonly assessed based on observations of fetal malformations and variations in rodent or rabbit studies. Such animal studies are costly, resource-intensive, and the results seen in one species often differ from those seen in other species or from those that might be relevant in humans. Some of the most promising non-animal testing alternatives make use of the self-organizing potential of embryonic stem cells to recapitulate developmental processes that may be sensitive to chemical exposure. The Stemina devTOXqP test uses human embryonic stem cells (hESCs) or human induced pluripotent stem cells (iPSCs) to predict developmental toxicity based on changes in cellular metabolism following drug or chemical exposure. The iPSC is a reprogrammed cell that is able to recapitulate development into all cell types like hESCs but does not come from an embryo.

In the EPA study, 1,065 ToxCast chemicals were first screened in single-concentration for the targeted biomarkers, the ratio of the amino acids ornithine (ORN) to cystine (CYSS), in response to the tested compound. Of the screened chemicals, 17% were predicted by the Stemina assay to cause developmental toxicity. These compounds were then tested at eight concentrations in the devTOXqP test to determine the exposure level at which the compound was considered to be toxic. The assay performance reached 82% accuracy with 67% sensitivity and 84% specificity. The sensitivity of the assay improved when more stringent evidence of toxicity was applied to the animal studies. Statistical analysis of the most potent chemical hits on specific biochemical targets in ToxCast provided insights into the mechanistic underpinnings of the targeted endpoint of the devTOXqP platform. The researchers found that an imbalance in Ornithine/Cystine was highly predictive of a chemicals potential to disrupt the development of an embryo or fetus, halting the pregnancy or producing birth defects.

The extensive nature of this research helps to define the applicability domain of the test in other words, where does it perform well and where will it need to be paired with other endpoints to generate a better understanding of the potential to cause birth defects, said Jessica Palmer, M.S., associate director of toxicology at Stemina. This is just the first step in our longer-term goal of moving away from a reliance on animal tests to predict human response and provides a foundation for building integrated testing systems focused on human cells.

To address concerns about the effects of drugs and chemicals on our health, we need more human-relevant methods to assess toxicity, said Kristie Sullivan, M.P.H., vice president for research policy at the Physicians Committee for Responsible Medicine. We encourage regulatory agencies and companies to consider how the devTOXqP test can improve safety and help reduce animal testing.

The publication titled, Profiling the ToxCast library with a pluripotent human (H9) stem cell line-based biomarker assay for developmental toxicity, can be accessed at: https://doi.org/10.1093/toxsci/kfaa014.

About EPA ToxCast Program

EPA ToxCast Program is developing approaches to predict chemical toxicity using data from high-throughput and high content in vitro assays. The goal of ToxCast is to develop and verify "toxicity signatures," which are algorithms using in vitro and in silico data to predict in vivo toxicities. ToxCast has data for approximately 1,800 chemicals from a broad range of sources including industrial and consumer products, environmental chemicals, and pharmaceutical compounds. More information available here.

About the Physicians Committee for Responsible Medicine

Founded in 1985, the Physicians Committee for Responsible Medicine is a nonprofit organization that promotes preventive medicine, conducts clinical research, and encourages higher standards for ethics and effectiveness in research. http://www.pcrm.org

About Stemina Biomarker Discovery, Inc.

Stemina Biomarker Discovery is a privately held company focused on the discovery, development and commercialization of molecular biomarkers to improve drug safety and human health. The companys cell-based assays use two innovative technologies: human stem cells and metabolomics. Stemina uses mass spectrometry to analyze the small molecules secreted by human stem cells in response to drugs or chemicals, injury, or disease. The company also uses differentiated human cells in its screens, like heart or neural cells made from human stem cells. The companys first commercial product, devTOX Discovery, launched in 2009, uses human stem cells to screen drug candidates, chemicals, consumer products and cosmetics for their potential effect on the developing human embryo. Subsequently launched drug screening programs, including devTOX quickPredict and Cardio quickPredict, continue the idea of bringing stem cell biology and metabolomics together for biomarker discovery and toxicity screening. For more information, please visit our website at http://www.stemina.com.

Originally posted here:
Stemina's Human Stem Cell-Based Test Demonstrates Potential to Predict Which Drugs and Chemicals Cause Birth Defects - Business Wire

Induced Pluripotent Stem Cells (iPSCs) Market Current Trends Strategies Involved, SWOT Analysis, Business Overview, Industry Challenges, and Forecast…

The Induced Pluripotent Stem Cells (iPSCs) MarketReport 2020 gives a clear understanding of the current market situation which includes of antique and projected upcoming market size based on technological growth, value and volume, projecting cost-effective and leading fundamentals in the Induced Pluripotent Stem Cells (iPSCs) market. Induced Pluripotent Stem Cells (iPSCs) industry report is to recognize, explain and forecast the global Induced Pluripotent Stem Cells (iPSCs) industry based on various aspects such as explanation, application, organization size, distribution mode, region. The Induced Pluripotent Stem Cells (iPSCs) Market report purposefully analyses every sub-segment regarding the individual growth trends, contribution to the total market, and the upcoming forecasts.

Global Induced Pluripotent Stem Cells (iPSCs) Market Segment by Type, covers

Global Induced Pluripotent Stem Cells (iPSCs) Market Segment by Applications, can be divided into

Get Free Exclusive Sample of this Premium Report at @ https://www.esherpamarketreports.com/request-sample/es-379889/

Global Induced Pluripotent Stem Cells (iPSCs) Market Segment by Manufacturers, this report covers:

Table of Contents1 Induced Pluripotent Stem Cells (iPSCs) Market Overview1.1 Product Overview and Scope of Induced Pluripotent Stem Cells (iPSCs)1.2 Induced Pluripotent Stem Cells (iPSCs) Segment by Type1.2.1 Global Induced Pluripotent Stem Cells (iPSCs) Production Growth Rate Comparison by Type 2020 VS 20261.2.2 Compact Type Induced Pluripotent Stem Cells (iPSCs)1.2.3 Standard Type Induced Pluripotent Stem Cells (iPSCs)1.3 Induced Pluripotent Stem Cells (iPSCs) Segment by Application1.3.1 Induced Pluripotent Stem Cells (iPSCs) Consumption Comparison by Application: 2020 VS 20261.4 Global Induced Pluripotent Stem Cells (iPSCs) Market by Region1.4.1 Global Induced Pluripotent Stem Cells (iPSCs) Market Size Estimates and Forecasts by Region: 2020 VS 20261.4.2 North America Estimates and Forecasts (2015-2026)1.4.3 Europe Estimates and Forecasts (2015-2026)1.4.4 China Estimates and Forecasts (2015-2026)1.4.5 Japan Estimates and Forecasts (2015-2026)1.5 Global Induced Pluripotent Stem Cells (iPSCs) Growth Prospects1.5.1 Global Induced Pluripotent Stem Cells (iPSCs) Revenue Estimates and Forecasts (2015-2026)1.5.2 Global Induced Pluripotent Stem Cells (iPSCs) Production Capacity Estimates and Forecasts (2015-2026)1.5.3 Global Induced Pluripotent Stem Cells (iPSCs) Production Estimates and Forecasts (2015-2026)2 Market Competition by Manufacturers2.1 Global Induced Pluripotent Stem Cells (iPSCs) Production Capacity Market Share by Manufacturers (2015-2020)2.2 Global Induced Pluripotent Stem Cells (iPSCs) Revenue Share by Manufacturers (2015-2020)2.3 Market Share by Company Type (Tier 1, Tier 2 and Tier 3)2.4 Global Induced Pluripotent Stem Cells (iPSCs) Average Price by Manufacturers (2015-2020)2.5 Manufacturers Induced Pluripotent Stem Cells (iPSCs) Production Sites, Area Served, Product Types2.6 Induced Pluripotent Stem Cells (iPSCs) Market Competitive Situation and Trends2.6.1 Induced Pluripotent Stem Cells (iPSCs) Market Concentration Rate2.6.2 Global Top 3 and Top 5 Players Market Share by Revenue2.6.3 Mergers & Acquisitions, Expansion3 Production Capacity by Region3.1 Global Production Capacity of Induced Pluripotent Stem Cells (iPSCs) Market Share by Regions (2015-2020)3.2 Global Induced Pluripotent Stem Cells (iPSCs) Revenue Market Share by Regions (2015-2020)3.3 Global Induced Pluripotent Stem Cells (iPSCs) Production Capacity, Revenue, Price and Gross Margin (2015-2020)3.4 North America Induced Pluripotent Stem Cells (iPSCs) Production3.4.1 North America Induced Pluripotent Stem Cells (iPSCs) Production Growth Rate (2015-2020)3.4.2 North America Induced Pluripotent Stem Cells (iPSCs) Production Capacity, Revenue, Price and Gross Margin (2015-2020)3.5 Europe Induced Pluripotent Stem Cells (iPSCs) Production3.5.1 Europe Induced Pluripotent Stem Cells (iPSCs) Production Growth Rate (2015-2020)3.5.2 Europe Induced Pluripotent Stem Cells (iPSCs) Production Capacity, Revenue, Price and Gross Margin (2015-2020)3.6 China Induced Pluripotent Stem Cells (iPSCs) Production3.6.1 China Induced Pluripotent Stem Cells (iPSCs) Production Growth Rate (2015-2020)3.6.2 China Induced Pluripotent Stem Cells (iPSCs) Production Capacity, Revenue, Price and Gross Margin (2015-2020)3.7 Japan Induced Pluripotent Stem Cells (iPSCs) Production3.7.1 Japan Induced Pluripotent Stem Cells (iPSCs) Production Growth Rate (2015-2020)3.7.2 Japan Induced Pluripotent Stem Cells (iPSCs) Production Capacity, Revenue, Price and Gross Margin (2015-2020)4 Global Induced Pluripotent Stem Cells (iPSCs) Consumption by Regions4.1 Global Induced Pluripotent Stem Cells (iPSCs) Consumption by Regions4.1.1 Global Induced Pluripotent Stem Cells (iPSCs) Consumption by Region4.1.2 Global Induced Pluripotent Stem Cells (iPSCs) Consumption Market Share by Region5 Production, Revenue, Price Trend by Type. And More

Click Here For Detailed Table Of Contents

Enquire before purchasing this report @ https://www.esherpamarketreports.com/pre-order-enquiry/es-379889

Reasons To Buy:

Purchase this Report with Full Access @ https://www.esherpamarketreports.com/purchase/es-379889/

Contact Us:

Name: Jason George

Email: [emailprotected]

Call :USA: +1 408 757 0510

Organization: eSherpa Market Reports

About Us:

eSherpa Market Reports is the credible source for gaining the market research reports that will exponentially accelerate your business. We are among the leading report resellers in the business world committed towards optimizing your business. The reports we provide are based on a research that covers a magnitude of factors such as technological evolution, economic shifts and a detailed study of market segment.

Original post:
Induced Pluripotent Stem Cells (iPSCs) Market Current Trends Strategies Involved, SWOT Analysis, Business Overview, Industry Challenges, and Forecast...

Biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation for functional sweat gland regeneration – Science…

Abstract

Mesenchymal stem cells (MSCs) encapsulation by three-dimensionally (3D) printed matrices were believed to provide a biomimetic microenvironment to drive differentiation into tissue-specific progeny, which made them a great therapeutic potential for regenerative medicine. Despite this potential, the underlying mechanisms of controlling cell fate in 3D microenvironments remained relatively unexplored. Here, we bioprinted a sweat gland (SG)like matrix to direct the conversion of MSC into functional SGs and facilitated SGs recovery in mice. By extracellular matrix differential protein expression analysis, we identified that CTHRC1 was a critical biochemical regulator for SG specification. Our findings showed that Hmox1 could respond to the 3D structure activation and also be involved in MSC differentiation. Using inhibition and activation assay, CTHRC1 and Hmox1 synergistically boosted SG gene expression profile. Together, these findings indicated that biochemical and structural cues served as two critical impacts of 3D-printed matrix on MSC fate decision into the glandular lineage and functional SG recovery.

Mesenchymal stem cells (MSCs) hold great promise for therapeutic tissue engineering and regenerative medicine, largely because of their capacity for self-renewal and multipotent properties (1). However, their uncertain fate has a major impact on their envisioned therapeutic use. Cell fate regulation requires specific transcription programs in response to environmental cues (2, 3). Once stem cells are removed from their microenvironment, their response to environmental cues, phenotype, and functionality could often be altered (4, 5). In contrast to growing information concerning transcriptional regulation, guidance from the extracellular matrix (ECM) governing MSC identity and fate determination is not well understood. It remains an active area of investigation and may provide previously unidentified avenues for MSC-based therapy.

Over the past decade, engineering three-dimensional (3D) ECM to direct MSC differentiation has demonstrated great potential of MSCs in regenerative medicine (6). 3D ECM has been found to be useful in providing both biochemical and biophysical cues and to stabilize newly formed tissues (7). Culturing cells in 3D ECM radically alters the interfacial interactions with the ECM as compared with 2D ECM, where cells are flattened and may lose their differentiated phenotype (8). However, one limitation of 3D materials as compared to 2D approaches was the lack of spatial control over chemistry with 3D materials. One possible solution to this limitation is 3D bioprinting, which could be used to design the custom scaffolds and tissues (9).

In contrast to traditional engineering techniques, 3D cell printing technology is especially advantageous because it can integrate multiple biophysical and biochemical cues spatially for cellular regulation and ensure complex structures with precise control and high reproducibility. In particular, for our final goal of clinical practice, extrusion-based bioprinting may be more appropriate for translational application. In addition, as a widely used bioink for extrusion bioprinting, alginate-based hydrogel could maintain stemness of MSC due to the bioinert property and improve biological activity and printability by combining gelatin (10).

Sweat glands (SGs) play a vital role in thermal regulation, and absent or malfunctioning SGs in a hot environment can lead to hyperthermia, stroke, and even death in mammals (11, 12). Each SG is a single tube consisting of a functionally distinctive duct and secretory portions. It has low regenerative potential in response to deep dermal injury, which poses a challenge for restitution of lost cells after wound (13). A major obstacle in SG regeneration, similar to the regeneration of most other glandular tissues, is the paucity of viable cells capable of regenerating multiple tissue phenotypes (12). Several reports have described SG regeneration in vitro; however, dynamic morphogenesis was not identified nor was the overall function of the formed tissues explored (1416). Recent advances in bioprinting and tissue engineering led to the complexities in the matrix design and fabrication with appropriate biochemical cues and biophysical guidance for SG regeneration (1719).

Here, we adopted 3D bioprinting technique to mimic the regenerative microenvironment that directed the specific SG differentiation of MSCs and ultimately guided the formation and function of glandular tissue. We used alginate/gelatin hydrogel as bioinks in this present study due to its good cytocompatibility, printability, and structural maintenance in long-time culture. Although the profound effects of ECM on cell differentiation was well recognized, the importance of biochemical and structural cues of 3D-printed matrix that determined the cell fate of MSCs remained unknown; thus, the present study demonstrated the role of 3D-printed matrix cues on cellular behavior and tissue morphogenesis and might help in developing strategies for MSC-based tissue regeneration or directing stem cell lineage specification by 3D bioprinting.

The procedure for printing the 3D MSC-loaded construct incorporating a specific SG ECM (mouse plantar region dermis, PD) was shown schematically in Fig. 1A. A 3D cellular construct with cross section 30 mm 30 mm and height of 3 mm was fabricated by using the optimized process parameter (20). The 3D construct demonstrated a macroporous grid structure with hydrogel fibers evenly distributed according to the computer design. Both the width of the fibers and the gap between the fibers were homogeneous, and MSCs were embedded uniformly in the hydrogel matrix fibers to result in a specific 3D microenvironment. (Fig. 1B).

(A) Schematic description of the approach. (B) Full view of the cellular construct and representative microscopic and fluorescent images and the quantitative parameters of 3D-printed construct (scale bars, 200 m). Photo credit: Bin Yao, Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA. (C) Representative microscopy images of cell aggregates and tissue morphology at 3, 7, and 14 days of culture (scale bars, 50 m) and scanning electron microscopy (sem) images of 3D structure (scale bars, 20 m). PD+/PD, 3D construct with and without PD. (D) DNA contents, collagen, and GAGs of native tissue and PD. (E) Proliferating cells were detected through Ki67 stain at 3, 7, and 14 days of culture. (F) Live/dead assay show cell viability at days 3, 7, and 14. *P < 0.05.

During the maintenance of constructs for stem cell expansion, MSCs proliferated to form aggregates of cells but self-assembled to an SG-like structure only with PD administration (Fig. 1C and fig. S1, A to C). We carried out DNA quantification assay to evaluate the cellular content in PD and found the cellular matrix with up to 90% reduction, only 3.4 0.7 ng of DNA per milligram tissue remaining in the ECM. We also estimated the proportions of collagen and glycosaminoglycans (GAGs) in ECM through hydroxyproline assay and dimethylmethylene blue assay, the collagen contents could increase to 112.6 11.3%, and GAGs were well retained to 81 9.6% (Fig. 1D). Encapsulated cells were viable, with negligible cell death apparent during extrusion and ink gelation by ionic cross-linking, persisting through extended culture in excess of 14 days. The fluorescence intensity of Ki67 of MSCs cultured in 2D condition decreased from days 3 (152.7 13.4) to 14 (29.4 12.9), while maintaining higher intensity of MSCs in 3D construct (such as 211.8 19.4 of PD+3D group and 209.1 22.1 of PD3D group at day 14). And the cell viability in 3D construct was found to be sufficiently high (>80%) when examined on days 3, 7, and 14. The phenomenon of cell aggregate formation and increased cell proliferation implied the excellent cell compatibility of the hydrogel-based construct and promotion of tissue development of 3D architectural guides, which did not depend on the presence or absence of PD (Fig. 1, E and F).

The capability of 3D-printed construct with PD directing MSC to SGs in vitro was investigated. The 3D construct was dissolved, and cells were isolated at days 3, 7, and 14 for transcriptional analysis. Expression of the SG markers K8 and K18 was higher from the 3D construct with (3D/PD+) than without PD (3D/PD); K8 and K18 expression in the 3D/PD construct was similar to with control that MSCs cultured in 2D condition, which implied the key role of PD in SG specification. As compared with the 2D culture condition, 3D administration (PD+) up-regulated SG markers, which indicated that the 3D structure synergistically boosted the MSC differentiation (Fig. 2A).

(A) Transcriptional expression of K8, K18, Fxyd2, Aqp5, and ATP1a1 in 3D-bioprinted cells with and without PD in days 3, 7, and 14 culture by quantitative real-time polymerase chain reaction (qRT-PCR). Data are means SEM. (B) Comparison of SG-specific markers K8 and K18 in 3D-bioprinted cells with and without PD (K8 and K18, red; DAPI, blue; scale bars, 50 m). (C and D) Comparison of SG secretion-related markers ATP1a1 (C) and Ca2+ (D) in 3D-bioprinted cells with and without PD [ATP1a1 and Ca2+, red; 4,6-diamidino-2-phenylindole (DAPI), blue; scale bars, 50 m].

In addition, we tested secretion-related genes to evaluate the function of induced SG cells (iSGCs). Although levels of the ion channel factors of Fxyd2 and ATP1a1 were increased notably in 2D culture with PD and ATP1a1 up-regulated in the 3D/PD construct, all the secretory genes of Fxyd2, ATP1a1, and water transporter Aqp5 showed the highest expression level in the 3D/PD+ construct (Fig. 2A). Considering the remarkable impact, further analysis focused on 3D constructs.

Immunofluorescence staining confirmed the progression of MSC differentiation. At day 7, cells in the 3D/PD+ construct began to express K8 and K18, which was increased at day 14, whereas cells in the 3D/PD construct did not express K8 and K18 all the time (Fig. 2B and fig. S2A). However, the expression of ATP1a1 (ATPase Na+/K+ transporting subunit alpha 1) and free Ca2+ concentration did not differ between cells in the 3D/PD+ and 3D/PD constructs (Fig. 2, C and D). By placing MSCs in such a 3D environment, secretion might be stimulated by rapid cell aggregation without the need for SG lineage differentiation. Cell aggregationimproved secretion might be due to the benefit of cell-cell contact (fig. S2B) (21, 22).

To map the cell fate changes during the differentiation between MSCs and SG cells, we monitored the mRNA levels of epithelial markers such as E-cadherin, occludin, Id2, and Mgat3 and mesenchymal markers N-cadherin, vimentin, Twist1, and Zeb2. The cells transitioned from a mesenchymal status to a typical epithelial-like status accompanied by mesenchymal-epithelial transition (MET), then epithelial-mesenchymal transition (EMT) occurred during the further differentiation of epithelial lineages to SG cells (fig. S3A). In addition, MET-related genes were dynamically regulated during the SG differentiation of MSCs. For example, the mesenchymal markers N-cadherin and vimentin were down-regulated from days 1 to 7, which suggested cells losing their mesenchymal phenotype, then were gradually up-regulated from days 7 to 10 in their response to the SG phenotype and decreased at day 14. The epithelial markers E-cadherin and occludin showed an opposite expression pattern: up-regulated from days 1 to 5, then down-regulated from days 7 to 10 and up-regulated again at day 14. The mesenchymal transcriptional factors ZEB2 and Twist1 and epithelial transcriptional factors Id2 and Mgat3 were also dynamically regulated.

We further analyzed the expression of these genes at the protein level by immunofluorescence staining (figs. S3B and S4). N-cadherin was down-regulated from days 3 to 7 and reestablished at day 14, whereas E-cadherin level was increased from days 3 to 7 and down-regulated at day 14. Together, these results indicated that a sequential and dynamic MET-EMT process underlie the differentiation of MSCs to an SG phenotype, perhaps driving differentiation more efficiently (23). However, the occurrence of the MET-EMT process did not depend on the presence of PD. Thus, a 3D structural factor might also participate in the MSC-specific differentiation (fig. S3C).

To investigate the underlying mechanism of biochemical cues in lineage-specific cell fate, we used quantitative proteomics analysis to screen the ECM factors differentially expressed between PD and dorsal region dermis (DD) because mice had eccrine SGs exclusively present in the pads of their paws, and the trunk skin lacks SGs. In total, quantitative proteomics analyses showed higher expression levels of 291 proteins in PD than DD. Overall, 66 were ECM factors: 23 were significantly up-regulated (>2-fold change in expression). We initially determined the level of proteins with the most significant difference after removing keratins and fibrin: collagen triple helix repeat containing 1 (CTHRC1) and thrombospondin 1 (TSP1) (fig. S5). Western blotting was performed to further confirm the expression level of CTHRC1 and TSP1, and we then confirmed that immunofluorescence staining at different developmental stages in mice revealed increased expression of CTHRC1 in PD with SG development but only slight expression in DD at postnatal day 28, while TSP1 was continuously expressed in DD and PD during development (Fig. 3, A to C). Therefore, TSP1 was required for the lineage-specific function during the differentiation in mice but was not dispensable for SG development.

(A and B) Differential expression of CTHRC1 and TSP1in PD and back dermis (DD) ECM of mice by proteomics analysis (A) and Western blotting (B). (C) CTHRC1 and TSP1 expression in back and plantar skin of mice at different developmental times. (Cthrc1/TSP1, red; DAPI, blue; scale bars, 50 m).

According to previous results of the changes of SG markers, 3D structure and PD were both critical to SG fate. Then, we focused on elucidating the mechanisms that underlie the significant differences observed in 2D and 3D conditions with or without PD treatment. To this end, we performed transcriptomics analysis of MSCs, MSCs treated with PD, MSCs cultured in 3D construct, and MSC cultured in 3D construct with PD after 3-day treatment. We noted that the expression profiles of MSCs treated with 3D, PD, or 3D/PD were distinct from the profiles of MSCs (Fig. 4A). Through Gene Ontology (GO) enrichment analysis of differentially expressed genes, it was shown that PD treatment in 2D condition induced up-regulation of ECM and inflammatory response term, and the top GO term for MSCs in 3D construct was ECM organization and extracellular structure organization. However, for the MSCs with 3D/PD treatment, we found very significant overrepresentation of GO term related to branching morphogenesis of an epithelial tube and morphogenesis of a branching structure, which suggested that 3D structure cues and biochemical cues synergistically initiate the branching of gland lineage (fig S6). Heat maps of differentially expressed ECM organization, cell division, gland morphogenesis, and branch morphogenesis-associated genes were shown in fig. S7. To find the specific genes response to 3D structure cues facilitating MSC reprogramming, we analyzed the differentially expressed genes of four groups of cells (Fig. 4B). The expression of Vwa1, Vsig1, and Hmox1 were only up-regulated with 3D structure stimulation, especially the expression of Hmox1 showed a most significant increase and even showed a higher expression addition with PD, which implied that Hmox1 might be the transcriptional driver of MSC differentiation response to 3D structure cues. Differential expression of several genes was confirmed by quantitative polymerase chain reaction (qPCR): Mmp9, Ptges, and Il10 were up-regulated in all the treated groups. Likewise, genes involving gland morphogenesis and branch morphogenesis such as Bmp2, Tgm2, and Sox9 showed higher expression in 3D/PD-treated group. Bmp2 was up-regulated only in 3D/PD-treated group, combined with the results of GO analysis, we assumed that Bmp2 initiated SG fate through inducing branch morphogenesis and gland differentiation (Fig. 4C).

(A) Gene expression file of four groups of cells (R2DC, MSCs; R2DT, MSC with PD treatment; R3DC, MSC cultured in 3D construct; and R3DT, MSC treated with 3D/PD). (B) Up-regulated genes after treatment (2DC, MSCs; 2DT, MSC with PD treatment; 3DC, MSC cultured in 3D construct; and 3DT, MSC treated with 3D/PD). (C) Differentially expressed genes were further validated by RT-PCR analysis. [For all RT-PCR analyses, gene expression was normalized to glyceraldehyde-3-phosphate dehydrogenase (GAPDH) with 40 cycles, data are represented as the means SEM, and n = 3].

To validate the role of HMOX1 and CTHRC1 in the differentiation of MSCs to SG lineages, we analyzed the gene expression of Bmp2 by regulating the expression of Hmox1 and CTHRC1 based on the 3D/PD-treated MSCs. The effects of caffeic acid phenethyl ester (CAPE) and tin protoporphyrin IX dichloride (Snpp) on the expression of Hmox1 were evaluated by quantitative real-time (qRT)PCR. Hmox1 expression was significantly activated by CAPE and reduced by Snpp. Concentration of CTHRC1 was increased with recombinant CTHRC1 and decreased with CTHRC1 antibody. That is, it was negligible of the effects of activator and inhibitor of Hmox1 and CTHRC1 on cell proliferation (fig. S8, A and B). Hmox1 inhibition or CTHRC1 neutralization could significantly reduce the expression of Bmp2, while Hmox1 activation or increased CTHRC1 both activated Bmp2 expression. Furthermore, Bmp2 showed highest expression by up-regulation of Hmox1 and CTHRC1 simultaneously and sharply decreased with down-regulation of Hmox1 and CTHRC1 at the same time (Fig. 5A). Immunofluorescent staining revealed that the expression of bone morphogenetic protein 2 (BMP2) at the translational level with CTHRC1 and Hmox1 regulation showed a similar trend with transcriptional changes (Fig. 5B). Likewise, the expression of K8 and K18 at transcriptional and translational level changed similarly with CTHRC1 and Hmox1 regulation (fig. S9, A and B). These results suggested that CTHRC1 and Hmox1 played an essential role in SG fate separately, and they synergistically induced SG direction from MSCs (Fig. 5C).

(A and B) Transcriptional analysis (A) and translational analysis (PD, MSCs; PD+, MSCs with 3D/PD treatment; CAPE, MSCs treated with 3D/PD and Hmox1 activator; Snpp, MSCs treated with 3D/PD and Hmox1 inhibitor; Cthrc1, MSCs treated with 3D/PD and recombinant CTHRC1; anti, MSCs treated with 3D/PD and CTHRC1 antibody: +/+, MSCs treated with 3D/PD and Hmox1 activator and recombinant CTHRC1; and /, MSCs treated with 3D/PD and Hmox1 inhibitor and CTHRC1 antibody. Data are represented as the means SEM and n = 3) (B) of bmp2 with regulation of CTHRC1 and Hmox1. (C) The graphic illustration of 3D-bioprinted matrix directed MSC differentiation. CTHRC1 is the main biochemical cues during SG development, and structural cues up-regulated the expression of Hmox1 synergistically initiated branching morphogenesis of SG. *P < 0.05.

Next, we sought to assess the repair capacity of iSGCs for in vivo implications, the 3D-printed construct with green fluorescent protein (GFP)labeled MSCs was transplanted in burned paws of mice (Fig. 6A). We measured the SG repair effects by iodine/starch-based sweat test at day 14. Only mice with 3D/PD treatment showed black dots on foot pads (representing sweating), and the number increased within 10 min; however, no black dots were observed on untreated and single MSC-transplanted mouse foot pads even after 15 min (Fig. 6B). Likewise, hematoxylin and eosin staining analysis revealed SG regeneration in 3D/PD-treated mice (Fig. 6C). GFP-positive cells were characterized as secretory lumen expressing K8, K18, and K19. Of note, the GFP-positive cells were highly distributed in K14-positive myoepithelial cells of SGs but were absent in K14-positive repaired epidermal wounds (Fig. 6, D and E). Thus, differentiated MSCs enabled directed restitution of damaged SG tissues both at the morphological and functional level.

(A) Schematic illustration of approaches for engineering iSGCs and transplantation. (B) Sweat test of mice treated with different cells. Photo credit: Bin Yao, Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Sciences, General Hospital of PLA. (C) Histology of plantar region without treatment and transplantation of MSCs and iSGCs (scale bars, 200 m). (D) Involvement of GFP-labeled iSGCs in directed regeneration of SG tissue in thermal-injured mouse model (K14, red; GFP, green; DAPI, blue; scale bar, 200 m). (E) SG-specific markers K14, K19, K8, and K18 detected in regenerated SG tissue (arrows). (K14, K19, K8, and K18, red; GFP, green; scale bars, 50 m).

A potential gap in MSC-based therapy still exists between current understandings of MSC performance in vivo in their microenvironment and their intractability outside of that microenvironment (24). To regulate MSCs differentiation into the right phenotype, an appropriate microenvironment should be created in a precisely controlled spatial and temporal manner (25). Recent advances in innovative technologies such as bioprinting have enabled the complexities in the matrix design and fabrication of regenerative microenvironments (26). Our findings demonstrated that directed differentiation of MSCs into SGs in a 3D-printed matrix both in vitro and in vivo was feasible. In contrast to conventional tissue-engineering strategies of SG regeneration, the present 3D-printing approach for SG regeneration with overall morphology and function offered a rapid and accurate approach that may represent a ready-to-use therapeutic tool.

Furthermore, bioprinting MSCs successfully repaired the damaged SG in vivo, suggesting that it can improve the regenerative potential of exogenous differentiated MSCs, thereby leading to translational applications. Notably, the GFP-labeled MSC-derived glandular cells were highly distributed in K14-positive myoepithelial cells of newly formed SGs but were absent in K14-positive repaired epidermal wounds. Compared with no black dots were observed on single MSC-transplanted mouse foot pads, the black dots (representing sweating function) can be observed throughout the entire examination period, and the number increased within 10 min on MSC-bioprinted mouse foot pads. Thus, differentiated MSCs by 3D bioprinting enabled exclusive restitution of damaged SG tissues morphologically and functionally.

Although several studies indicated that engineering 3D microenvironments enabled better control of stem cell fates and effective regeneration of functional tissues (2730), there were no studies concerning the establishment of 3D-bioprinted microenvironments that can preferentially induce MSCs differentiating into glandular cells with multiple tissue phenotypes and overall functional tissue. To find an optimal microenvironment for promoting MSC differentiation into specialized progeny, biochemical properties are considered as the first parameter to ensure SG specification. In this study, we used mouse PD as the main composition of a tissue-specific ECM. As expected, this 3D-printed PD+ microenvironment drove the MSC fate decision to enhance the SG phenotypic profile of the differentiated cells. By ECM differential protein expression analysis, we identified that CTHRC1 was a critical biochemical regulator of 3D-printed matrix for SG specification. TSP1 was required for the lineage-specific function during the differentiation in mice but was not dispensable for SG development. Thus, we identified CTHRC1 as a specific factor during SG development. To our knowledge, this is the first demonstration of CTHRC1 involvement in dictating MSC differentiation to SG, highlighting a potential therapeutic tool for SG injury.

The 3D-printed matrix also provided architectural guides for further SG morphogenesis. Our results clearly show that the 3D spatial dimensionality allows for better cell proliferation and aggregation and affect the characteristics of phenotypic marker expression. Notably, the importance of 3D structural cues on MSC differentiation was further proved by MET-EMT process during differentiation, where the influences did not depend on the presence of biochemical cues. To fully elucidate the underlying mechanisms, we first examined how 3D structure regulating stem cell fate choices. According to our data, Hmox1 is highly up-regulated in 3D construct, which were supposed to response to hypoxia, with a previously documented role in MSC differentiation (31, 32). It is suggested that 3D microenvironment induced rapid cell aggregation leading to hypoxia and then activated the expression of Hmox1.

Through regulation of the expression of Hmox1 and addition or of CTHRC1 in the matrix, we confirmed that each of them is critical for SG reprogramming, respectively. Thus, biochemical and structural cues of 3D-printed matrix synergistically creating a microenvironment could enhance the accuracy and efficiency of MSC differentiation, thereby leading to resulting SG formation. Although we further need a more extensive study examining the role of other multiple cues and their possible overlap function in regulating MSC differentiation, our findings suggest that CTHRC1 and Hmox1 provide important signals that cooperatively modulate MSC lineage specification toward sweat glandular lineage. The 3D structure combined with PD stimulated the GO functional item of branch morphogenesis and gland formation, which might be induce by up-regulation of Bmp2 based on the verification of qPCR results. Although our results could not rule out the involvement of other factors and their possible overlapping role in regulating MSC lineage specification toward SGs, our findings together with several literatures suggested that BMP2 plays a critical role in inducing branch morphogenesis and gland formation (3335).

In summary, our findings represented a novel strategy of directing MSC differentiation for functional SG regeneration by using 3D bioprinting and pave the way for a potential therapeutic tool for other complex glandular tissues as well as further investigation into directed differentiation in 3D conditions. Specifically, we showed that biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation, and our results highlighted the importance of 3D-printed matrix cues as regulators of MSC fate decisions. This avenue opens up the intriguing possibility of shifting from genetic to microenvironmental manipulations of cell fate, which would be of particular interest for clinical applications of MSC-based therapies.

The main aim and design of the study was first to determine whether by using 3D-printed microenvironments, MSCs can be directed to differentiate and regenerate SGs both morphologically and functionally. Then, to investigate the underlying molecular mechanism of biochemical and structural cues of 3D-printed matrix involved in MSCs reprogramming. The primary aims of the study design were as follows: (i) cell aggregation and proliferation in a 3D-bioprinted construct; (ii) differentiation of MSCs at the cellular phenotype and functional levels in the 3D-bioprinted construct; (iii) the MET-EMT process during differentiation; (iv) differential protein expression of the SG niche in mice; (v) differential genes expression of MSCs in 3D-bioprinted construct; (vi) the key role of CTHRC1 and HMOX1 in MSCs reprogramming to SGCs; and (vii) functional properties of regenerated SG in vivo.

Gelatin (Sigma-Aldrich, USA) and sodium alginate (Sigma-Aldrich, USA) were dissolved in phosphate-buffered saline (PBS) at 15 and 1% (w/v), respectively. Both solutions were sterilized under 70C for 30 min three times at an interval of 30 min. The sterilized solutions were packed into 50-ml centrifuge tubes, stored at 4C, and incubated at 37C before use.

From wild-type C57/B16 mice (Huafukang Co., Beijing) aged 5 days old, dermal homogenates were prepared by homogenizing freshly collected hairless mouse PD with isotonic phosphate buffer (pH 7.4) for 20 min in an ice bath to obtain 25% (w/v) tissue suspension. The supernatant was obtained after centrifugation at 4C for 20 min at 10,000g. The DNA content was determined using Hoechst 33258 assay (Beyotime, Beijing). The fluorescence intensity was measured to assess the amount of remaining DNA within the decellularized ECMs and the native tissue using a fluorescence spectrophotometer (Thermo Scientific, Evolution 260 Bio, USA). The GAGs content was estimated via 1,9-dimethylmethylene blue solution staining. The absorbance was measured with microplate reader at wavelength of 492 nm. The standard curve was made using chondroitin sulfate A. The total COL (Collagen) content was determined via hydroxyproline assay. The absorbance of the samples was measured at 550 nm and quantified by referring to a standard curve made with hydroxyproline.

MSCs were bioprinted with matrix materials by using an extrusion-based 3D bioprinter (Regenovo Co., Bio-Architect PRO, Hangzhou). Briefly, 10 ml of gelatin solution (10% w/v) and 5 ml of alginate solution (2% w/v) were warmed under 37C for 20 min, gently mixed as bioink and used within 30 min. MSCs were collected from 100-mm dishes, dispersed into single cells, and 200 l of cell suspension was gently mixed with matrix material under room temperature with cell density 1 million ml1. PD (58 g/ml) was then gently mixed with bioink. Petri dishes at 60 mm were used as collecting plates in the 3D bioprinting process. Within a temperature-controlled chamber of the bioprinter, with temperature set within the gelation region of gelatin, the mixture of MSCs and matrix materials was bioprinted into a cylindrical construct layer by layer. The nozzle-insulation temperature and printing chamber temperature were set at 18 and 10C, respectively; nozzles with an inner diameter of 260 m were chosen for printing. The diameter of the cylindrical construct was 30 mm, with six layers in height. After the temperature-controlled bioprinting process, the printed 3D constructs were immersed in 100-mM calcium chloride (Sigma-Aldrich, USA) for 3 min for cross-linking, then washed with Dulbeccos modified Eagle medium (DMEM) (Gibco, USA) medium for three times. The whole printing process was finished in 10 min. The 3D cross-linked construct was cultured in DMEM in an atmosphere of 5% CO2 at 37C. The culture medium was changed to SG medium [contains 50% DMEM (Gibco, New York, NY) and 50% F12 (Gibco) supplemented with 5% fetal calf serum (Gibco), 1 ml/100 ml penicillin-streptomycin solution, 2 ng/ml liothyronine sodium (Gibco), 0.4 g/ml hydrocortisone succinate (Gibco), 10 ng/ml epidermal growth factor (PeproTech, Rocky Hill, NJ), and 1 ml/100 ml insulin-transferrin-selenium (Gibco)] 2 days later. The cell morphology was examined and recorded under an optical microscope (Olympus, CX40, Japan).

Fluorescent live/dead staining was used to determine cell viability in the 3D cell-loaded constructs according to the manufacturers instructions (Sigma-Aldrich, USA). Briefly, samples were gently washed in PBS three times. An amount of 1 M calcein acetoxymethyl (calcein AM) ester (Sigma-Aldrich, USA) and 2 M propidium iodide (Sigma-Aldrich, USA) was used to stain live cells (green) and dead cells (red) for 15 min while avoiding light. A laser scanning confocal microscopy system (Leica, TCSSP8, Germany) was used for image acquisition.

The cell-printed structure was harvested and fixed with a solution of 4% paraformaldehyde. The structure was embedded in optimal cutting temperature (OCT) compound (Sigma-Aldrich, USA) and sectioned 10-mm thick by using a cryotome (Leica, CM1950, Germany). The sliced samples were washed repeatedly with PBS solution to remove OCT compound and then permeabilized with a solution of 0.1% Triton X-100 (Sigma-Aldrich, USA) in PBS for 5 min. To reduce nonspecific background, sections were treated with 0.2% bovine serum albumin (Sigma-Aldrich, USA) solution in PBS for 20 min. To visualize iSGCs, sections were incubated with primary antibody overnight at 4C for anti-K8 (1:300), anti-K14 (1:300), anti-K18 (1:300), anti-K19 (1:300), anti-ATP1a1 (1:300), anti-Ki67 (1:300), antiN-cadherin (1:300), antiE-cadherin (1:300), anti-CTHRC1 (1:300), or anti-TSP1 (1:300; all Abcam, UK) and then incubated with secondary antibody for 2 hours at room temperature: Alexa Fluor 594 goat anti-rabbit (1:300), fluorescein isothiocyanate (FITC) goat anti-rabbit (1:300), FITC goat anti-mouse (1:300), or Alexa Fluor 594 goat anti-mouse (1:300; all Invitrogen, CA). Sections were also stained with 4,6-diamidino-2-phenylindole (Beyotime, Beijing) for 15 min. Stained samples were visualized, and images were captured under a confocal microscope.

To harvest the cells in the construct, the 3D constructs were dissolved by adding 55 mM sodium citrate and 20 mM EDTA (Sigma-Aldrich, USA) in 150 mM sodium chloride (Sigma-Aldrich, USA) for 5 min while gently shaking the petri dish for better dissolving. After transfer to 15-ml centrifuge tubes, the cell suspensions were centrifuged at 200 rpm for 3 min, and the supernatant liquid was removed to harvest cells for further analysis.

Total RNA was isolated from cells by using TRIzol reagent (Invitrogen, USA) following the manufacturers protocol. RNA concentration was measured by using a NanoPhotometer (Implen GmbH, P-330-31, Germany). Reverse transcription involved use of a complementary DNA synthesis kit (Takara, China). Gene expression was analyzed quantitatively by using SYBR green with the 7500 Real-Time PCR System (Takara, China). The primers and probes for genes were designed on the basis of published gene sequences (table S1) (National Center for Biotechnology Information and PubMed). The expression of each gene was normalized to that for glyceraldehyde-3-phosphate dehydrogenase and analyzed by the 2-CT method. Each sample was assessed in triplicate.

The culture medium was changed to SG medium with 2 mM CaCl2 for at least 24 hours, and cells were loaded with fluo-3/AM (Invitrogen, CA) at a final concentration of 5 M for 30 min at room temperature. After three washes with calcium-free PBS, 10 M acetylcholine (Sigma-Aldrich, USA) was added to cells. The change in the Fluo 3 fluorescent signal was recorded under a laser scanning confocal microscopy.

Cell proliferation was evaluated through CCK-8 (Cell counting kit-8) assay. Briefly, cells were seeded in 96-well plates at the appropriate concentration and cultured at 37C in an incubator for 4 hours. When cells were adhered, 10 l of CCK-8 working buffer was added into the 96-well plates and incubated at 37C for 1 hour. Absorbance at 450 nm was measured with a microplate reader (Tecan, SPARK 10M, Austria).

Proteomics of mouse PD and DD involved use of isobaric tags for relative and absolute quantification (iTRAQ) in BGI Company, with differentially expressed proteins detected in PD versus DD. Twofold greater difference in expression was considered significant for further study.

Tissues were grinded and lysed in radioimmunoprecipitation assay buffer (Beyotime, Nanjing). Proteins were separated by 12% SDSpolyacrylamide gel electrophoresis and transferred to a methanol-activated polyvinylidene difluoride membrane (GE Healthcare, USA). The membrane was blocked for 1 hour in PBS with Tween 20 containing 5% bovine serum albumin (Sigma-Aldrich, USA) and probed with the antibodies anti-CTHRC1 (1:1000) and anti-TSP1 (1:1000; both Abcam, UK) overnight at 4C. After 2 hours of incubation with goat anti-rabbit horseradish peroxidaseconjugated secondary antibody (Santa Cruz Biotechnology, CA), the protein bands were detected by using luminal reagent (GE Healthcare, ImageQuant LAS 4000, USA).

Total RNA was prepared with TRIzol (Invitrogen), and RNA sequencing was performed using HiSeq 2500 (Illumina). Genes with false discovery rate < 0.05, fold difference > 2.0, and mean log intensity > 2.0 were considered to be significant.

CAPE or Snpp was gently mixed with bioink at a concentration of 10 M. Physiological concentration of CTHRC1 was measured by enzyme linked immunosorbent assay (ELISA) (80 ng/ml), and then recombinant CTHRC1 or CTHRC1 antibody was added into the bioink at a concentration of 0.4 g/ml. The effect of inhibitor and activator was estimated by qRT-PCR or ELISA.

Mice were anesthetized with pentobarbital (100 mg/kg) and received subcutaneous buprenorphine (0.1 mg/kg) preoperatively. Full-thickness scald injuries were created on paw pads with soldering station (Weller, WSD81, Germany). Mice recovered in clean cages with paper bedding to prevent irritation or infection. Mice were monitored daily and euthanized at 30 days after wounding. Mice were maintained in an Association for Assessment and Accreditation of Laboratory Animal Careaccredited animal facility, and procedures were performed with Institutional Animal Care and Use Committeeapproved protocols.

MSCs in 3D-printed constructs with PD were cultured with DMEM for 2 days and then replaced with SG medium. The SG medium was changed every 2 days, and cells were harvested on day 12. The K18+ iSGCs were sorting through flow cytometry and injected into the paw pads (1 106 cells/50 l) of the mouse burn model by using Microliter syringes (Hamilton, 7655-01, USA). Then, mice were euthanized after 14 days; feet were excised and fixed with 10% formalin (Sigma-Aldrich, USA) overnight for paraffin sections and immunohistological analysis.

The foot pads of anesthetized treated mice were first painted with 2% (w/v) iodine/ethanol solution then with starch/castor oil solution (1 g/ml) (Sigma-Aldrich, USA). After drying, 50 l of 100 M acetylcholine (Sigma-Aldrich, USA) was injected subcutaneously into paws of mice. Pictures of the mouse foot pads were taken after 5, 10, and 15 min.

All data were presented as means SEM. Statistical analyses were performed using GraphPad Prism7 statistical software (GraphPad, USA). Significant differences were calculated by analysis of variance (ANOVA), followed by the Bonferroni test when performing multiple comparisons between groups. P < 0.05 was considered as a statistically significant difference.

Supplementary material for this article is available at http://advances.sciencemag.org/cgi/content/full/6/10/eaaz1094/DC1

Fig. S1. Biocompatibility of 3D-bioprinted construct and cellular morphology in 2D monolayer culture.

Fig. S2. Expression of SG-specific and secretion-related markers in MSCs and SG cells in vitro.

Fig. S3. Transcriptional and translational expression of epithelial and mesenchymal markers in 3D-bioprinted cells with and without PD.

Fig. S4. Expression of N- and E-cadherin in MSCs and SG cells in 2D monolayer culture.

Fig. S5. Proteomic microarray assay of differential gene expression between PD and DD ECM in postnatal mice.

Fig. S6. GO term analysis of differentially expressed pathways.

Fig. S7. Heat maps illustrating differential expression of genes implicated in ECM organization, cell division, and gland and branch morphogenesis.

Fig. S8. The expression of Hmox1 and the concentration of CTHRC1 on treatment and the related effects on cell proliferation.

Fig. S9. The expression of K8 and K18 with Hmox1 and CTHRC1 regulation.

Table S1. Primers for qRT-PCR of all the genes.

This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial license, which permits use, distribution, and reproduction in any medium, so long as the resultant use is not for commercial advantage and provided the original work is properly cited.

Acknowledgments: Funding: This study was supported in part by the National Nature Science Foundation of China (81571909, 81701906, 81830064, and 81721092), the National Key Research Development Plan (2017YFC1103300), Military Logistics Research Key Project (AWS17J005), and Fostering Funds of Chinese PLA General Hospital for National Distinguished Young Scholar Science Fund (2017-JQPY-002). Author contributions: B.Y. and S.H. were responsible for the design and primary technical process, conducted the experiments, collected and analyzed data, and wrote the manuscript. Y.W. and R.W. helped perform the main experiments. Y.Z. and T.H. participated in the 3D printing. W.S. and Z.L. participated in cell experiments and postexamination. S.H. and X.F. collectively oversaw the collection of data and data interpretation and revised the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data needed to evaluate the conclusions in the paper are present in the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors.

Originally posted here:
Biochemical and structural cues of 3D-printed matrix synergistically direct MSC differentiation for functional sweat gland regeneration - Science...

With Over 280 Therapies Under Evaluation, the Stem Cell Therapy Market is Estimated to be Worth USD 8.5 Billion by 2030, Claims Roots Analysis – P&T…

The success of approved stem cell therapies has caused a surge in interest of biopharma developers in this field; many innovator companies are currently progressing proprietary leads across different phases of clinical development, with cautious optimism

LONDON, March 4, 2020 /PRNewswire/ -- Roots Analysishas announced the addition of "Global Stem Cells Market: Focus on Clinical Therapies, 20202030 (Based on Source (Allogeneic, Autologous); Origin (Adult, Embryonic); Type (Hematopoietic, Mesenchymal, Progenitor); Lineage (Amniotic Fluid, Adipose Tissue, Bone Marrow, Cardiosphere, Chondrocytes, Corneal Tissue, Cord Blood, Dental Pulp, Neural Tissue Placenta, Peripheral Blood, Stromal Cells); and Potency (Multipotent, Pluripotent))" report to its list of offerings.

There is a growing body of evidence supporting the vast applicability and superiority of treatment outcomes of stem cell therapies, compared to conventional treatment options. In fact, the unmet needs within this domain have spurred the establishment of many start-ups in recent years.

To order this 500+ page report, which features 185+ figures and 220+ tables, please visit this link

Key Market Insights

Over 280 stem cell therapies are under development, most of which are allogeneic products

More than 50% of the pipeline candidates are in the mid to late phase trials (phase II and above), and allogenic therapies (majority of which are derived from the bone marrow) make up 65% of the pipeline.

70% of pipeline candidates are based on mesenchymal stem cells

It is worth highlighting that the abovementioned therapies are designed to treat musculoskeletal (22%), neurological (21%) and cardiovascular (15%) disorders. On the other hand, hematopoietic stem cell-based products are mostly being evaluated for the treatment of oncological disorders, primarily hematological malignancies.

Close to 85% stem cell therapy developers are based in North America and Asia-Pacific regions

Within these regions, the US, China, South Korea and Japan, have emerged as key R&D hubs for stem cell therapies. It is worth noting that majority of the initiatives in this domain are driven by small / mid-sized companies

Over 1,500 grants were awarded for stem cell research, since 2015

More than 45% of the total amount was awarded under the R01 mechanism (which supports research projects). The NCI, NHLBI, NICHD, NIDDK, NIGMS and OD emerged as key organizations that have offered financial support for time periods exceeding 25 years as well.

Outsourcing has become indispensable to R&D and manufacturing activity in this domain

Presently, more than 80 industry / non-industry players, based in different regions across the globe, claim to provide contract development and manufacturing services to cater to the unmet needs of therapy developers. Examples include (in alphabetical order) Bio Elpida, Cell and Gene Therapy Catapult, Cell Tech Pharmed, GenCure, KBI Biopharma, Lonza, MEDINET, Nikon CeLL innovation, Roslin Cell Therapies, WuXi Advanced Therapies and YposKesi.

North America and Asia-Pacific markets are anticipated to capture over 80% share by 2030

The stem cell therapies market is anticipated to witness an annualized growth rate of over 30% during the next decade. Interestingly, the market in China / broader Asia-Pacific region is anticipated to grow at a relatively faster rate.

To request a sample copy / brochure of this report, please visit this link

Key Questions Answered

The USD 8.5 billion (by 2030) financial opportunity within the stem cell therapies market has been analyzed across the following segments:

The report features inputs from eminent industry stakeholders, according to whom stem cell therapies are currently considered to be a promising alternatives for the treatment of a myriad of disease indications, with the potential to overcome challenges associated with conventional treatment options. The report includes detailed transcripts of discussions held with the following experts:

The research covers brief profiles of several companies (including those listed below); each profile features an overview of the company, financial information (if available), stem cell therapy portfolio and an informed future outlook.

For additional details, please visit

https://www.rootsanalysis.com/reports/view_document/stem-cells-market/296.htmlor email sales@rootsanalysis.com

You may also be interested in the following titles:

Contact:Gaurav Chaudhary+1(415)800-3415+44(122)391-1091Gaurav.Chaudhary@rootsanalysis.com

Logo: https://mma.prnewswire.com/media/742223/Roots_Analysis_Logo.jpg

Read more:
With Over 280 Therapies Under Evaluation, the Stem Cell Therapy Market is Estimated to be Worth USD 8.5 Billion by 2030, Claims Roots Analysis - P&T...