Category Archives: Stem Cell Treatment


Looking to the future with Dr. Francis Collins – UAB News

In a talk at UAB on March 6, the NIH director shared his thoughts on exceptional opportunities for science and young scientists and highlighted several exciting UAB projects.

NIH Director Francis Collins, M.D., Ph.D., visited UAB on March 6. In addition to his public talk, Collins had breakfast with UAB medical students and met with groups of young researchers and other investigators across campus.Speaking to a packed University of Alabama at Birmingham audience March 6, Francis Collins, M.D., Ph.D., director of the National Institutes of Health, shared his picks of 10 areas of particular excitement and promise in biomedical research.

In nearly every area, UAB scientists are helping to lead the way as Collins himself noted in several cases. At the conclusion of his talk, Collins addedhis advice for young scientists. Here is Collins top 10 list, annotated with some of the UAB work ongoing in each area and ways that faculty, staff and students can get involved.

I am so jazzed with what has become possible with the ability to study single cells and see what they are doing, Collins said. They have been out of our reach now we have reached in. Whether you are studying rheumatoid arthritis, diabetes or the brain, you have the chance to ask each cell what it is doing.

Single-cell sequencing and UAB:Collins noted that Robert Carter, M.D., the acting director of the National Institute of Arthritis and Musculoskeletal and Skin Diseases, was a longtime faculty member at UAB (serving as director of the Division of Clinical Immunology and Rheumatology). For the past several years, UAB researchers have been studying gene expression in subpopulations of immune cells inpatients with rheumatoid arthritis.

Join in:Researchers can take advantage of the single-cell sequencing core facility in UABsComprehensive Flow Cytometry Core, directed by John Mountz, M.D., Ph.D., Goodwin-Blackburn Research Chair in Immunology and professor in the Department of Medicine Division of Clinical Immunology and Rheumatology.

Learn more:Mountz and other heavy users of single-cell sequencing explain how the techniqueslet them travel back in time and morein this UAB Reporter story.

The NIHsBRAIN Initiativeis making this the era where we are going to figure out how the brain works all 86 billion neurons between your ears, Collins said. The linchpin of this advance will be the development of tools to identify new brain cell types and circuits that will improve diagnosis, treatment and prevention of autism, schizophrenia, Parkinsons and other neurological conditions, he said.

Brain tech and UAB:Collins highlighted thework of BRAIN Initiative granteeHarrison Walker, M.D., an associate professor in the Department of Neurology, whose lab has been developing a more sophisticated way to understand the benefits of deep brain stimulation for people with Parkinsons and maybe other conditions, Collins said.

Join in:UABs planned new doctoral program in neuroengineering would be the first of its kind in the country.

Learn more:Find out why neuroengineering is asmart career choicein this UAB Reporter story.

Researchers can now take a blood cell or skin cell and, by adding four magic genes, Collins explained, induce the cells to become stem cells. These induced pluripotent stem (iPS) cells can then in turn be differentiated into any number of different cell types, including nerve cells, heart muscle cells or pancreatic beta cells. The NIH has invested in technology to put iPS-derived cells on specialized tissue chips. Youve got you on a chip, Collins explained. Some of us dream of a day where this might be the best way to figure out whether a drug intervention is going to work for you or youre going to be one of those people that has a bad consequence.

iPS cells at UAB:Collins displayed images of thecutting-edge cardiac tissue chipdeveloped by a UAB team led by Palaniappan Sethu, Ph.D., an associate professor in the Department of Biomedical Engineering and the Division of Cardiovascular Disease. The work allows the development of cardiomyocytes that can be used to study heart failure and other conditions, Collins said.

Join in:UABs biomedical engineering department, one of the leading recipients of NIH funding nationally, is a joint department of the School of Engineering and School of Medicine. Learn more about UABsundergraduate and graduate programs in biomedical engineering, and potential careers, here.

Learn more:See howthis novel bioprinterdeveloped by UAB biomedical researchers is speeding up tissue engineering in this story from UAB News.

We have kind of ignored the fact that we have all these microbes living on us and in us until fairly recently, Collins said. But now it is clear that we are not an organism we are a superorganism formed with the trillions of microbes present in and on our bodies, he said. This microbiome plays a significant role not just in skin and intestinal diseases but much more broadly.

Microbiome at UAB:Collins explained that work led by Casey Morrow, Ph.D., and Casey Weaver, M.D., co-directors of theMicrobiome/Gnotobiotics Shared Facility, has revealed intriguing information abouthow antibiotics affect the gut microbiome. Their approach has potential implications for understanding, preserving and improving health, Collins said.

Join in:Several ongoing clinical trials at UAB are studying the microbiome, including a studymodifying diet to improve gut microbiotaand an investigation of the microbiomes ofpostmenopausal women looking for outcomes and response to estrogen therapy.

Learn more:This UAB News storyexplains the UAB researchthat Collins highlighted.

Another deadly influenza outbreak is likely in the future, Collins said. What we need is not an influenza vaccine that you have to redesign every year, but something that would actually block influenza viruses, he said. Is that even possible? It just might be.

Influenza research at UAB:Were probably at least a decade away from a universal influenza vaccine. But work ongoing at UAB in the NIH-fundedAntiviral Drug Discovery and Development Center(AD3C), led by Distinguished Professor Richard Whitley, M.D., is focused on such an influenza breakthrough.

Join in:For now, the most important thing you can do to stop the flu is to get a flu vaccination. Employees can schedule afree flu vaccination here.

Learn more:Why get the flu shot? What is it like? How can you disinfect your home after the flu? Get all the information atthis comprehensive sitefrom UAB News.

The NIH has a role to play in tackling the crisis of opioid addiction and deaths, Collins said. The NIHs Helping to End Addiction Long-term (HEAL) initiative is an all-hands-on-deck effort, he said, involving almost every NIH institute and center, with the goal of uncovering new targets for preventing addiction and improving pain treatment by developing non-addictive pain medicines.

Addiction prevention at UAB:A big part of this initiative involves education to help professionals and the public understand what to do, Collins said. The NIH Centers of Excellence in Pain Education (CoEPE), including one at UAB, are hubs for the development, evaluation and distribution of pain-management curriculum resources to enhance pain education for health care professionals.

Join in:Find out how to tell if you or a loved one has a substance or alcohol use problem, connect with classes and resources or schedule an individualized assessment and treatment through theUAB Medicine Addiction Recovery Program.

Learn more:Discover some of the many ways that UAB faculty and staff aremaking an impact on the opioid crisisin this story from UAB News.

We are all pretty darn jazzed about whats happened in the past few years in terms of developing a new modality for treating cancer we had surgery, we had radiation, we had chemotherapy, but now weve got immunotherapy, Collins said.

Educating immune system cells to go after cancer in therapies such as CAR-T cell therapy is the hottest science in cancer, he said. I would argue this is a really exciting moment where the oncologists and the immunologists together are doing amazing things.

Immunotherapy at UAB:I had to say something about immunology since Im at UAB given that Max Cooper, whojust got the Lasker Awardfor [his] B and T cell discoveries, was here, Collins said. This is a place I would hope where lots of interesting ideas are going to continue to emerge.

Join in:The ONeal Comprehensive Cancer Center at UAB is participating in a number of clinical trials of immunotherapies.Search the latest trials at the Cancer Centerhere.

Learn more:Luciano Costa, M.D., Ph.D., medical director of clinical trials at the ONeal Cancer Center, discusses the promise ofCAR-T cell therapy in this UAB MedCast podcast.

Assistant Professor Ben Larimer, Ph.D., is pursuing a new kind of PET imaging test that could give clinicians afast, accurate picture of whether immunotherapy is workingfor a patient in this UAB Reporter article.

The All of Us Research Program from NIH aims to enroll a million Americans to move away from the one-size-fits-all approach to medicine and really understand individual differences, Collins said. The program, which launched in 2018 and is already one-third of the way to its enrollment goal, has a prevention rather than a disease treatment approach; it is collecting information on environmental exposures, health practices, diet, exercise and more, in addition to genetics, from those participants.

All of Us at UAB:UAB has been doing a fantastic job of enrolling participants, Collins noted. In fact, the Southern Network of the All of Us Research Program, led by UAB, has consistently been at the top in terms of nationwide enrollment, as School of Medicine Dean Selwyn Vickers, M.D., noted in introducing Collins.

Join in:Sign up forAll of Usat UAB today.

Learn more:UABs success in enrolling participants has led to anew pilot study aimed at increasing participant retention rates.

Rare Disease Day, on Feb. 29, brought together hundreds of rare disease research advocates at the NIH, Collins said. NIH needs to play a special role because many diseases are so rare that pharmaceutical companies will not focus on them, he said. We need to find answers that are scalable, so you dont have to come up with a strategy for all 6,500 rare diseases.

Rare diseases at UAB: The Undiagnosed Diseases Network, which includes aUAB siteled by Chief Genomics Officer Bruce Korf, M.D., Ph.D., is a national network that brings together experts in a wide range of conditions to help patients, Collins said.

Participants in theAlabama Genomic Health Initiative, also led by Korf, donate a small blood sample that is tested for the presence of specific genetic variants. Individuals with indications of genetic disease receive whole-genome sequencing. Collins noted that lessons from the AGHI helped guide development of the All of Us Research Program.

Collins also credited UABs Tim Townes, Ph.D., professor emeritus in the Department of Biochemistry and Molecular Genetics, for developing the most significantly accurate model of sickle cell disease in a mouse which has been a great service to the [research] community. UAB is now participating in anexciting clinical trial of a gene-editing technique to treat sickle cellalong with other new targeted therapies for the devastating blood disease.

Join in:In addition to UABs Undiagnosed Diseases Program (which requires a physician referral) and the AGHI, patients and providers can contact theUAB Precision Medicine Institute, led by Director Matt Might, Ph.D. The institute develops precisely targeted treatments based on a patients unique genetic makeup.

Learn more:Discover how UAB experts solved medical puzzles for patients by uncovering anever-before-described mutationandcracking a vomiting mysteryin these UAB News stories.

We know that science, like everything else, is more productive when teams are diverse than if they are all looking the same, Collins said. My number one priority as NIH director is to be sure we are doing everything we can to nurture and encourage the best and brightest to join this effort.

Research diversity at UAB:TheNeuroscience Roadmap Scholars Programat UAB, supported by an NIH R25 grant, is designed to enhance engagement and retention of under-represented graduate trainees in the neuroscience workforce. This is one of several UAB initiatives to increased under-represented groups and celebrate diversity. These include several programs from theMinority Health and Health Disparities Research Centerthat support minority students from the undergraduate level to postdocs; thePartnership Research Summer Training Program, which provides undergraduates and especially minority students with the opportunity to work in UAB cancer research labs; theDeans Excellence Award in Diversityin the School of Medicine; and the newly announcedUnderrepresented in Medicine Senior Scholarship Programfor fourth-year medical students.

Join in:The Roadmap program engages career coaches and peer-to-peer mentors to support scholars. To volunteer your expertise, contact Madison Bamman atmdbamman@uab.eduorvisit the program site.

Learn more:Farah Lubin, Ph.D., associate professor in the Department of Neurobiology and co-director of the Roadmap Scholars Program,shares the words and deeds that can save science careersin this Reporter story. In another story, Upender Manne, Ph.D., professor in the Department of Pathology and a senior scientist in the ONeal Comprehensive Cancer Center, explains how students in the Partnership Research Summer Training Program gethooked on cancer research.

In answer to a students question, Collins also shared his advice to young scientists. One suggestion: Every investigator needs to be pretty comfortable with some of the computational approaches to science, Collins said. Big data is here artificial intelligence, machine-learning. We can all get into that space. But its going to take some training, and it will be really helpful to have those skills.

Join in:UAB launched aMaster of Science in Data Scienceprogram in fall 2018.

Learn more:Discover how UAB researchers areusing machine-learning in their labsand toimprove cancer treatment. Those looking for a free introduction cantake advantage of the Data Science Clubfrom UAB IT Research Computing.

Go here to read the rest:
Looking to the future with Dr. Francis Collins - UAB News

Single-cell mass cytometry reveals cross-talk between inflammation-dampening and inflammation-amplifying cells in osteoarthritic cartilage – Science…

INTRODUCTION

Osteoarthritis (OA) is a highly prevalent, age-related disease of the joints, characterized by cartilage degeneration, loss of mobility, and chronic pain. Much work has been done investigating several aspects of its complex etiology, including the contributions of metabolic, epigenetic, genetic, and cellular factors. However, no disease-modifying drugs exist to treat OA, with the current standard of care being limited to pain management, followed by eventual joint replacement. Recent and ongoing work has highlighted the important interplay between aging, inflammation, and loss of regenerative potential in multiple tissues. Although cartilage is a relatively simple tissue, with a single cell type being encapsulated in its secreted extracellular matrix, the variable degree of degeneration associated with each patient with OA suggests that understanding this tissue at a single-cell level can provide insights into the onset and progression of pathology.

Defining the precise subpopulations that constitute cartilage will also aid strategies for cartilage tissue engineering or for enhancing endogenous cartilage regeneration. Unlike other skeletal tissues, cartilage has a remarkably low regeneration potential. Even injuries sustained in youth remain unrepaired, giving rise to the fibrocartilaginous tissue that can lead to accelerated OA pathology. Multiple studies have explored the putative cartilage progenitor cells (CPCs) in articular cartilage by characterizing their cell surface markers and describing their function (1). Notably, the CPC populations were reported to be enriched in OA cartilage, having an increased migratory potential, the ability to form highly clonal populations, and multipotency (i.e., the ability to give rise to chondrocytes, osteoblasts, and adipocytes in culture) (24). Recently, the human skeletal stem cell (hSSC) was identified (5), further suggesting another fountain of cells for repair. However, despite the existence of these putative regenerative populations, overall cartilage repair remains low, both in healthy and diseased states. Cartilage repair is variable even in younger, non-OA patients who undergo cartilage related injuries, such as anterior cruciate ligament rupture or degenerative meniscal tears, with some patients having a good recovery while others develop OA over a decade or so. Collectively, this suggests that there are factors preventing effective repair and regeneration of the tissue and that these factors vary between patients.

One source of this limited repair might be the chronic inflammation experienced by the joint. The synovium is known to be infiltrated by a variety of immune cells (6), and several inflammatory cytokines have been detected in the synovial fluid of patients with OA (7). Further, several studies have characterized the actions of the hypoxia factors (HIFs), nitric oxide, reactive oxygen species, nuclear factor B (NF-B) signaling, and other pathways that maintain the proinflammatory environment. To understand how this milieu might affect the proregenerative populations, such as the CPCs, we used single-cell mass cytometry [cytometry by time-of-flight (cyTOF)] to map both the proregenerative cell populations and inflammatory populations. By simultaneously being able to map cell identity and signaling states, we could observe how cells interact and influence each other. Furthermore, these maps provide us with a cell populationbased stratification of patients with OA, which may aid in targeted OA therapeutics in the future.

Toward our goal of profiling rare stem/progenitor-like populations within normal and OA cartilage, we used cyTOF, a mass spectrometrybased high-dimensional method for single-cell detection of isotope-labeled antibodies (Fig. 1A) (8). While cyTOF panels have to be preselected for each experiment, this technique provides the advantage that a large number of cells can be easily profiled in multiple samples without being cost prohibitive. This profiling at the protein level is complementary to single-cell transcriptomics and can provide a snapshot of the active signaling pathways in a specific subpopulation. After a detailed study of the literature and our preliminary data, a panel of 33 markers was labeled and optimized (see Methods and table S1) for profiling chondrocytes. This panel included cell surface receptors, adhesion molecules, signaling mediators, and cell cycle and transcription factors that are known to be important for cartilage homeostasis (table S1). Samples were collected from the surgical waste of patients with OA undergoing total knee arthroplasty [according to an Institutional Review Board (IRB) protocol approved by Stanford University], digested and expanded for a single passage in high-density culture as previously described (9). Each sample had an expression ratio of Col2a1/Col1a1 between 10 and 100 (fig. S1A), and the expression of MMP3, MMP9, and MMP13 was 10- to 10,000-fold higher in OA cartilage compared to normal, as expected (fig. S1, B to D). An average of 3 104 and 10 104 cells were assayed per OA or normal sample, respectively, and, to ensure chondrogenicity, only the SOX99/CD44 double-positive cells were further analyzed (fig. S1E). For visualization, the total population was downsampled to 9%, representing 9000 cells, and cells were projected onto a two-dimensional plane using t-distributed stochastic neighbor embedding (tSNE) (Fig. 1B). The spatial representations of OA and normal cells are distinct, although no single sample (patient) for either normal or OA samples was observed to dominate this representation (Fig. 1, C and D). Analysis of SOX9 and CD44 staining showed high levels of staining across all cells, ensuring the chondrogenic phenotype of the cells with no dedifferentiation observed during sample processing (Fig. 1, E and F). We proceeded to analyze the single-cell data from 20 OA and 5 normal samples. We observed known features of the OA landscape, for example, the expansion of NOTCH1-expressing chondrocytes in OA (Fig. 1, E and F). Phosphorylated NF-B (pNF-B), in contrast, could not readily distinguish between normal and OA samples, which both consisted of populations manifesting high, medium, and low levels of signaling (Fig. 1, E and F).

(A) Schematic outlining the procedures used to profile chondrocytes by mass cytometry. Briefly, cells are dissociated from cartilage tissue, stained with metal-conjugated antibodies, and analyzed using cyTOF. The resulting data are then gated for live, SOX9/CD44-positive chondrocytes that are used for downstream analyses, including identifying clusters with FlowSOM. (B) tSNE projections of the normal (blue) and OA (red) chondrocytes where each cell is represented by a dot. Each group was downsampled randomly to 9000 cells. (C) Normal chondrocytes colored by patient sample, downsampled to 9000 cells. (D) OA chondrocytes colored by patient sample, downsampled to 9000 cells. (E) tSNE plots of 9000 normal chondrocytes, colored by the expression of two chondrogenic markers (SOX9 and CD44), the cell surface receptor NOTCH1, and pNF-B. Expression is set at the max of each channel and is comparable between (E) and (F). (F) tSNE plots of 9000 OA chondrocytes, colored by the expression of two chondrogenic markers (SOX9 and CD44), the cell surface receptor NOTCH1, and pNF-B. Expression is set at the max of each channel and is comparable between (E) and (F).

To find unique subpopulations in the normal and OA cartilage, we used the algorithm FlowSOM (10) to define clusters (see Methods) based on the similarity of expression of cell surface receptors and intracellular markers. FlowSOM identified 20 clusters or subpopulations in our data (Fig. 2, A and B ). Using an alternate algorithm, X-shift (11), we observed a similar number and composition of clusters (fig. S1F), providing an independent validation of the FlowSOM analyses. A standard scaled distribution matrix for all the surface receptors and intracellular markers used to define the 20 clusters with FlowSOM (Fig. 2C) demonstrates the molecular identity of these clusters. For example, clusters 1 and 2 are marked by high intercellular adhesion molecule (ICAM), clusters 12 and 16 have a high expression of NOTCH1, STRO1, and CD166, and clusters 15 and 20 have high interleukin-1 receptor 1 (IL1R1) and tumor necrosis factor receptor II (TNFRII). Using the 20 clusters, we observed that the patients with OA were highly anticorrelated with the normal samples (fig. S2A). On the basis of the known functions of the molecules that defined each subpopulation, we broadly defined clusters as CPC clusters or non-CPC clusters (Fig. 2D and fig. S2B).

(A) Abundance of each of the 20 clusters called by FlowSOM analysis in normal samples. Each point represents a single sample. (n = 5). (B) Abundance of each of the 20 clusters called by FlowSOM analysis in OA samples (n = 20). Each point represents a single sample. (C) Expression of cell surface receptors used for delineating the 20 clusters. Expression is averaged between all cells of a given cluster ID. Color is scaled to 1 for each protein between all the clusters. Dendograms were drawn using complete-linkage hierarchical clustering. (D) Table of the cluster IDs that are enriched, depleted, or similar between OA and normal samples. Colors in the enriched section correspond to the tSNE projection on the right. The tSNE projection contains cells from clusters that are enriched in OA compared to normal, sampled to 9000 cells. Enrichment, depletion, or similarity between the ranked means of normal (n = 5) and OA (n = 20) cluster abundance was tested using an unpaired, two-tailed Mann-Whitney test with Bonferroni correction ( = 0.0025). Adjusted P values for all enriched or depleted clusters are 0.002. (E) Coefficient of variation (mean divided by SD) for each cluster in normal or OA samples. (F) Shannons diversity index (H) calculated for each normal and OA sample (see Methods). Theoretical max H value is 2.99. Equality between the means H values for OA (n = 20) and normal (n = 5) samples was tested using a two-tailed Mann-Whitney test. ***P = 0.001. (G) Hierarchical clustering of normal and OA samples by cluster abundances. Abundance is scaled to 1. Samples belonging to the three designated groups are labeled at the bottom. (H) Average cluster abundance in normal and group A, B, and C patients with OA. Each color designates a cluster ID.

We next wanted to investigate how the nature and frequency of the identified subpopulations varied between the normal and OA samples, specifically to determine whether populations were gained or lost with disease. On the basis of this idea, we categorized the clusters into three groups: (i) increased in OA, (ii) unchanged between OA and normal, and (iii) decreased in OA. Eight subpopulations (clusters 5, 7, 9, 11, 12, 13, 19, and 20) were enriched in the OA samples compared to normal; five subpopulations (clusters 1, 2, 3, 8, and 14) were depleted compared to normal, while seven subpopulations (clusters 4, 6, 10, 15, 16, 17, and 18) remained unchanged between the OA and normal samples (Fig. 2D and fig. S2B). Quantitation of the frequency of these populations revealed interpatient heterogeneity, which we quantified using the coefficient of variation (Fig. 2E). CPC clusters 4 and 16 along with the non-CPC cluster 19 were among the most variable between patients with OA, while clusters 15 and 20 were the least variable (Fig. 2E). As an alternative way to quantify this heterogeneity, we used a metric used in population ecology, known as Shannons diversity index, which describes how heterogeneous and evenly distributed populations are in an ecosystem. On the basis of the 20 populations identified by FlowSOM, we observed that (i) OA samples had a higher Shannon diversity index (H value) and, additionally, (ii) the range of H values for patients with OA was larger than for normal samples, indicating a loss of population evenness in OA (Fig. 2F). A direct comparison between the OA and normal samples is difficult, however, as the number of OA samples (n = 20) is much larger than normal samples (n = 5). Hence, this dataset may be missing some of the potentially higher variability clusters in normal cartilage.

Using these populations, whose unique identities are detailed in later sections, we performed hierarchical clustering of the 20 OA patient and 5 normal samples in our study. Our goal was to identify subsets of patients with unique compositions of these rare populations. Such characterization, common in the cancer field, can be helpful in designing targeted therapeutic strategies tailored to groups of patients with similar molecular underpinnings driving their disease. As expected, all the normal samples clustered together (Fig. 2G). We observed three major groups of patients, with some patients that clustered only with themselves. Group A, the largest of the three groups with 12 patients, was enriched in clusters 7 and 11, marked by CD105 expression (Fig. 2, G and H). Group B, consisting of three patients, was enriched in clusters 17 and 18, the CD24+ populations, and group C, also consisting of three patients, was characterized by a high abundance of clusters 9, 12, and 16 that were identified to be NOTCH1/VCAM-1 (vascular cell adhesion molecule1)positive CPC (Fig. 2, G and H). In the following sections, we will detail the unique characteristics of these populations and the etiology that they reveal about the underlying patients with OA.

Several studies (3, 4, 1219) have found CPCs that have the ability to give rise to chondrocytes, show self-renewal in culture, and are able to migrate in OA cartilage. These CPCs are believed to be the origin of the highly clonal characteristic clusters (20, 21) found in OA cartilage. Their role in OA disease pathology, however, remains unclear, especially whether they contribute to disease onset and progression. To address these questions and better characterize the CPCs and their cross-talk with other cartilage-resident cells, we had designed our cyTOF panel to include 13 previously described markers for CPCs (Fig. 3A and table S1). Of the 20 clusters identified using FlowSOM, 12 clusters were found to be enriched for these CPC markers in a variety of combinations (Fig. 3A and fig. S3A). The rest of the clusters, designated non-CPC, are very low in their expression of the CPC markers as shown for clusters 3, 5, and 6 (fig. S2A). In contrast to previous observations, we found that there are three variants of CPC subpopulations that are depleted in OA (Fig. 3B), which we termed CPC I. Out of the rest, two clusters were unchanged between normal and OA cartilage, termed CPC II, and six clusters were enriched in OA cartilage, comprising some of the previously described CPC populations, which we termed CPC III (Fig. 3B).

(A) Expression of the 13 CPC markers among the clusters that are enriched for them. Expression is scaled to 1 between all clusters. (B) tSNE projections of the type I (depleted), type II (similar), and type III (enriched) CPCs in OA, colored by cluster ID, where each cluster ID has a different color. Cells are sampled to 9000 when possible. (C) Cell cycle analysis for each cluster. Cell cycle stages were analyzed for each cell, and then, the proportion of the population in G0 and in the cell cycle was calculated for each cluster. The percentage in the cell cycle is given to the right of each bar graph. (D) Cell signaling and other intracellular and cell surface receptor markers for the CPC clusters. Expression is scaled to 1. (E) Cluster abundance for each sample in the OA groups and normal cells. Significance is tested with a multiple-test corrected Welchs t test. ns, not significant. (F) Correlation between abundance of each cluster, labeled on each axis. Each point represents a patient with OA. The full matrix of correlations between clusters is plotted in fig. S3A. *P = 0.05, **P = 0.01, and ***P = 0.001.

The CPC I clusters were characterized by lower CD105 expression in contrast to the CPC III clusters (Fig. 3A). Cluster 1 and 2 cells were distinct in having a high expression of CD54 (ICAM) (Fig. 3A). Previous work exploring markers for stem or progenitor cells had noted that cells with high CD54 and CD55 expression had higher levels of aldehyde dehydrogenase activity, associated with stem cell function (22). Cluster 14 was distinguished by the expression of CD151, i.e., tetraspanin, a cell adhesion marker, which was described to mark chondrocytes with higher chondrogenic potential in an in vitro study (23). Cell cycle analysis showed that CPC I clusters had the highest percentage of cells that were cycling (Fig. 3C), although, overall, the number of cycling cells was low, as expected for postmitotic chondrocytes (<20%). The CPC I clusters are exclusively characterized by extracellular signalregulated kinase 1/2 signaling, while the other clusters, with the exception of the CPC II cluster 10, are not (Fig. 3D). Out of the CPC II clusters, cluster 4 is characterized by a high CD73 expression and is not predominantly active in any of the tested signaling pathways (Fig. 3D). CD73 has recently been identified to be one of the critical markers on an adult hSSC population (5). The CPC III populations contained clusters that were enriched for many inflammatory signaling pathways. Clusters 12, 13, and 16 were high in the expression of pNF-B, pSTAT3 (phosphorylated Signal transducer and activator of transcription 3), -catenin, and HIF2A, associated with inflammation in OA. However, CPC III also had populations that were low in these pathways, namely, clusters 7, 9, and 11 (Fig. 3D). Cluster 16 appears to be the quintessential CD105/CD90-high, NOTCH1/STRO1driven migratory CPC that has been previously identified in OA cartilage (15, 24). Group C patients had a significantly higher percentage of the proinflammatory clusters 9, 12, and 16 and a lower percentage of low-inflammation clusters 7 and 11 (Fig. 3E). This anticorrelation between clusters 9 and 11, clusters 12 and 7, and clusters 16 and 14 (Fig. 3F and fig. S3, A and B) held across the 20-patient cohort, suggesting that these patients might be particularly driven by this cellular subtype.

We further analyzed the non-CPC populations that were identified by our panel, with a focus on putative inflammatory populations that might contribute to pathology. Among these were clusters 15 and 20, which are characterized by the coexpression of two cytokine receptors, IL1R1 (CD121A) and TNFRII (CD120B) (Fig. 4, A to C). Cluster 20 is significantly expanded in OA cartilage compared to the normal cartilage (Fig. 4D). Clusters 15 and 20 vary in the quantity IL1R1 expression, with cluster 20 having a higher level of IL1R1 (Fig. 4E). However, both clusters 15 and 20 have similarly high levels of TNFRII and HIF2A expression (Fig. 4E).

(A) tSNE projection of normal cells (gray) with clusters 15 and 20 colored, sampled at 9000 cells. (B) tSNE projection of OA cells (gray) with clusters 15 and 20 colored, sampled at 9000 cells. (C) A magnified projection of clusters 15 and 20 from normal and OA samples, ****P = 0.0001. (D) Quantification of the abundance of clusters 15 and 20 in normal and OA samples. Significance is tested using Welchs t test. Each point represents a sample. (E) Magnified projection of clusters 15 and 20 depicting expression of the two cell surface receptors, TNFRII and IL1R1, and of intracellular HIF2A. Expression is scaled to max value in dataset for each protein and is comparable across normal and OA samples. Heatmap below the tSNE depicts quantification of average expression in representative chondrocytes (cluster 5) in comparison to clusters 15 and 20. (F) Single-cell RNA (scRNA) sequencing data from (25) reanalyzed. Cells expressing TNFRII and IL1R1 were sorted in silico, and their transcriptome was compared to the rest of the OA cells and used for Gene Ontology (GO) term and STRING analyses. (G) Same as in (E), for signaling markers pJNK1/2, pNF-B (H), and pSMAD1/5 (I). (J) Fold change in cytokines from human 62-plex Luminex assay between dimethyl sulfoxide (DMSO) and JNK inhibitor treatment. (K) Fold change in cytokines from human 62-plex Luminex assay between DMSO and NF-B inhibitor treatment. (L) Fold change in cytokines from human 62-plex Luminex assay between DMSO and Alk inhibitor treatment. (M) Raw mean fluorescence intensity (MFI) values for cytokines that were significantly altered between DMSO- and JNK-treated samples in at least five of six tested OA samples. Significance was first tested for using analysis of variance (ANOVA) with multiple corrections for the 62 comparisons, and then, t test with Tukeys correction was applied for each comparison on a patient-by-patient sample. Each point represents an independent technical treatment and cytokine analyses for the same patient (n = 6 patients with OA). AU, arbitrary units. (N and O) Same as in (M) but with NF-B and Alk inhibitors, respectively (n = 3 patients with OA). *P = 0.05, **P = 0.01, and ***P = 0.001.

To further understand the molecular underpinnings of these subpopulations, we used publicly available single-cell RNA (scRNA) sequencing data (25). We were able to successfully identify cells that expressed both IL1R1 and TNFRSF1B transcripts in the scRNA sequencing data. These cells represented about ~2% of sequenced cells, validating the frequency we observed by cyTOF. Chondrocytes that expressed both transcripts were sorted in silico, and the differentially expressed genes and pathways were analyzed. The IL1RI/TNFRII-expressing chondrocytes were found to be highly enriched in pathways related to innate and adaptive immune cells, inflammation, and altered T and B cells signaling in arthritis (Fig. 4F). These analyses suggest that the IL1RI/TNFRII cells might act to recruit immune cells to the joint space. We, therefore, termed clusters 15 and 20 inflammation-amplifying (Inf-A) chondrocytes. Upon analyzing their signaling status, the Inf-A clusters showed exclusive signaling through pJNK (phosphorylated c-Jun N-terminal kinase) and pSMAD1/5 compared to the rest of the chondrocyte clusters (Fig. 4, G and I). In contrast, pNF-B levels in clusters 15 and 20 were similar to other clusters identified (Fig. 4H). Despite its rarity, cluster 20 was highly consistent among patients, with TNFRII expression and JNK and SMAD1/5 phosphorylation levels consistently high across all patients with OA in cluster 20, and more variable in cluster 15 (fig. S4A). Cluster 20 shows the lowest coefficient of variation in the OA samples (Fig. 2E).

Next, we sought to explore the functional effects of inhibiting these Inf-A cells in OA cartilage by capitalizing on their distinct signaling through JNK. Chondrocytes derived from six patients were cultured for 48 hours in the presence of JNK inhibitor II, and the secretome was analyzed via 62 antibody human Luminex panels. Across all six patients, a variety of cytokines were altered (fig. S4B), many trending toward significance. Restricting our analysis to only those cytokines that were altered in five or more patients (>83% response rate), we observed a significant decrease in C-C motif chemokine ligand 2 (CCL2) and CCL7 after JNK inhibition (Fig. 4, J and M). CCL2 and CCL7 are well-established chemoattractants for monocytes and are known to be altered during OA progression (26). Genetic deletions of CCL2 and its receptor CCR2 prevent the development of surgical OA, further underscoring the importance of CCL2 as a key modulator in pathology (27). In contrast, inhibition of NF-B activity with BMS-345541 (28) did not affect CCL2 or CCL7 secretion in OA chondrocytes (Fig. 4, K and N), suggesting that the effect is specific to the Inf-A population. As a complementary approach, we inhibited SMAD1/5, the other exclusive signaling pathway of the Inf-A cells, using an ALK (activin receptor-like kinase) inhibitor. ALK receptors are the most common upstream target of SMAD1/5 signaling in OA (29). As hypothesized, ALK inhibitor treatment resulted in a decrease in the same cytokines affected by the JNK inhibitor, CCL2, and CCL7, and, additionally, C-X-C motif chemokine ligand 1 (CXCL1) and CXCL5 (Fig. 4, L and O), two other leukocyte attracting factors. Collectively, these data are consistent with the transcriptional data suggesting that the IFNR1 (interferon receptor 1)/TNFRIIcoexpressing cells mark a rare OA subpopulation that is potentially responsible for immune recruitment to the joint. We demonstrated that inhibition of this rare population can significantly affect the overall secretome of the end-stage OA chondrocytes.

Our previous work established a role for the cell surface receptor CD24 in mitigating inflammation in healthy and induced pluripotent stem cell (iPSC)-derived chondrocytes (30). Although CD24 is highly expressed in juvenile and iPSC-derived chondrocytes, its expression is decreased with age (30), potentially underscoring the age-related etiology of OA. We included CD24 in our cyTOF panel to understand the interplay of CD24+ cells with the other regenerative and inflammatory subpopulations in the OA joint. FlowSOM-derived clusters 17 and 18 were found to be most enriched in CD24 expression (Fig. 5, A and C). Both clusters 17 and 18 were found in equal numbers in normal and OA cartilage; however, there was a high variability in their abundance between patients (Fig. 5B). In agreement with our previous report, CD24 cells decreased with age (fig. S5A) and were among the least reactive groups to undergo stimulation by the proinflammatory cytokine IL1B (fig. S5B). Therefore, we termed clusters 17 and 18 inflammation-dampening (Inf-D) I and II cells, respectively. Inf-D II cells had the highest levels of CD24 expression and also had higher levels of Sox9 and CD44, although expression in Inf-D I cells was comparable with normal cells (Fig. 5C). To further characterize the function of these CD24+ cells, we used the same previously published scRNA sequencing dataset and sorted out CD24+ cells. Consistent with our hypothesis that the CD24+ cells are capable of immune modulation, we observed an enrichment for pathways related to inflammation and immune cell trafficking and cross-talk (Fig. 5D and fig. S5C). In addition, the CD24+ cells showed an enrichment of oxidative phosphorylation pathways, suggesting that these cells could have different metabolic processes compared to other chondrocytes (Fig. 5D and fig. S5C).

(A) tSNE projection of normal and OA cells (gray) with clusters 17 and 18 colored, sampled at 9000 cells each. (B) Abundance of each cluster per sample. Differences between the means were tested using Welchs t test. (C) Heatmaps of chondrogenic markers SOX9 and CD44, as well as CD24. Expression is scaled to the highest expressing cell in the group. (D) scRNA sequencing data from (25), reanalyzed. Cells expressing CD24 with a high Col2a1/Col1a1 ratio were sorted in silico, and their transcriptome was compared to the rest of the OA cells and used for GO term and STRING analyses. (E) Hierarchical clustering of OA samples based on clusters 15, 17, 18, and 20. Abundance is scaled to one for each cluster. Groups are labeled along the x axis. (F) Violin plots of abundance of clusters 17, 18, 15, and 20 in low and high Inf-D groups. Each sample is represented as a point. (G) Correlation between the abundance of cluster 20 with clusters 17 + 18. Ninety-five percent confidence interval is shown in gray dashed line. Slope of line tested is significantly nonzero. (H) tSNE projection of OA cells, with clusters 15, 20, and 19 labeled, sampled at 9000 cells. (I) Heatmaps of the average expression of each marker in the given cluster. (J) Fold change in cytokines from human 62-plex Luminex assay between control and 3-isobutyl-1-methylxanthine (IBMX) treatment. (K) Fold change in cytokines from human 62-plex Luminex assay between control and a combined IBMX and JNK inhibitor treatment. (L) Percent change in cytokine MFI between control and the combined IBMX/JNK inhibitor treatment.

To understand the interplay between Inf-A and Inf-D cells in the OA cartilage, we analyzed their abundance in the cohort of 20 patients and used hierarchical clustering to order patients by the content of their Inf-A and Inf-D cells. The patients were clearly stratified into two large categories of patients: Inf-Dlow and Inf-Dhigh patients with OA (Fig. 5E). The Inf-Dhigh group had concomitantly high levels of the Inf-A clusters than the Inf-Dlow group (Fig. 5F). In addition, a positive correlation was observed between the percentage of Inf-A and Inf-D cells in patients (Fig. 5G). This led us to hypothesize that a combination strategy of enhancing Inf-D while inhibiting Inf-A populations could be effective in mitigating inflammation in OA cartilage. We also noted a small and highly variable population, cluster 19, which had a mixed character. Cluster 19 showed IL1R1 expression without the inflammatory signature that we observed in the Inf-A I and Inf-A II cells (pJNK1/2 and pSMAD1/5) (Fig. 5, H and I ) and curiously also expressed CD24. These cells were only present in 8 of the 20 patients (Fig. 5I) but further suggested that CD24 expression in the Inf-D cells can dampen inflammation.

To test this hypothesis, we first induced mild CD24 overexpression by treating cells with 3-isobutyl-1-methylxanthine (IBMX), an adenosine 3,5-monophosphate inhibitor that has been shown to increase CD24 expression in adipocytes (31). Treatment with 0.5 mM IBMX for 48 hours up-regulated CD24 expression by two- to fourfold in OA chondrocytes (fig. S5D). IBMX increased the gene expression of the mitochondrial genes Tfam, and Pgc1a (fig. S5E), although no consistent effect was, however, observed on MMP13 expression (fig. S5E). Using the 62-plex Luminex assay, we observed a modest down-regulation of CCL2 and CCL7; however, these effects were milder than the direct inhibition of the Inf-A signaling (Fig. 5J).

We then tested a combination treatment of JNK inhibitor with IBMX for 48 hours. We observed a greater magnitude decreased in CCL2 and CCL7 with the combination treatment (Fig. 5K) as compared to the single treatment with JNK inhibitor (Fig. 4, J and M). In addition, the combination therapy further mitigated inflammation by reducing the secretion of targets such as IL21, IL22, VCAM, and IFNB1 (Fig. 5L). Similar to JNK inhibitor treatment, matrix metalloproteinase (MMP) gene expression remained unaffected by the combination treatment (fig. S5F). These data, however, suggest that targeting multiple combinations of rare cell types in OA cartilage may be beneficial in mitigating inflammation.

In this study, we built the first single-cell, proteomic atlas for healthy and osteoarthritic adult articular cartilage. Cartilage regeneration and OA remain unmet medical needs. Therefore, a high-resolution cellular atlas of articular cartilage tissue lays the foundation for insight into disease pathology, drug strategies, and tissue engineering. Using a panel of 33 markers, we identified multiple populations that constitute the articular cartilage landscape, including rare populations that contribute to disease pathology and interpatient heterogeneity.

Recently, an scRNA sequencing map of knee cartilage was reported from a cohort of 10 patients with OA and outlined several known and cell populations (25). Our study complements this single-cell transcriptomic data, with the additional advantage that the proteomic snapshot provides insight into the status of signaling pathways in the identified subpopulations. The single-cell proteomic approach is especially pertinent in robustly identifying rare cell populations that are difficult to discern from RNA sequencing data, where only 1600 cells were studied from all the patients with OA. In contrast, the ability to map 30,000 to 100,000 cells per patient in a 20-patient cohort by the cyTOF method provided us a robust dataset to find and validate statistically significant rare subpopulations. A recent study on rare senescent cell populations in OA cartilage has shown the influence of these small populations in OA pathology (32). Removal of senescent cells significantly impaired OA progression in a mouse model and modulated end-stage human OA chondrocytes, underscoring the need for further studies on other rare populations that might contribute to OA pathology. In addition, frequent discrepancies between gene and protein expression have been reported in OA, further signifying the need for complementary proteomic and transcriptomic studies.

The ability to measure a large number of cells with high precision allowed us to identify two, rare chondrocyte subpopulations (Inf-A and Inf-D), which constitute only 0.5 to 1.5% of all chondrocytes. However, pharmacologically targeting these small populations led to a dampening of inflammatory cytokines at the population level. The Inf-A cells express both the TNFRII and IL1R1 receptors, are consistently expanded in OA compared to normal cartilage, and are characterized by activated JNK1/2 and SMAD1/5 pathways. An analysis of their transcriptomes from the published scRNA sequencing dataset suggests that these cells may function to recruit immune cells. Inhibition of these cells using a JNK inhibitor led to an overall reduction of secreted CCL2 and CCL7, cytokines implicated in immune cell recruitment (33, 34). Genetic knockout of JNK1 or JNK2 ameliorates disease symptoms in a collagenase-induced model of rheumatoid arthritis (RA) (35), and inhibition of JNK protects joints from characteristic degeneration (36). These mouse models can be used in future studies to test a putative immune cell recruitment function of the Inf-A population. However, unlike in RA models, JNK inhibitors have not been systematically studied as a therapy in animal models of OA. TNFRII antibodies also have a strong therapeutic index in RA (37). Our work suggests that some of these therapies may also be successful in targeting OA.

The other novel population identified in our study is the Inf-D chondrocytes, which are characterized by the expression of CD24, a cell surface receptor we had previously reported to be enriched in juvenile cartilage and associated with resistance to inflammatory cues (30). Expression of CD24 in Inf-A cells, a subpopulation observed in some patients, led to complete inhibition of JNK activation. In addition, the positive correlation between Inf-A and Inf-D populations in a subset of patients led us to hypothesize an interplay between these two populations. Combinatorial treatment with JNK inhibitor (lowering Inf-D) and IBMX, a small molecular activator of CD24 (increasing Inf-D), showed a greater decrease in CCL2, CCL7, CXCL1, CXCL5, and other inflammatory cytokines than JNK inhibition alone. Our data, therefore, provide insights into the interplay between multiple cellular populations that likely contribute to the chronic inflammatory environment that is observed in end-stage OA cartilage. A deeper understanding of these populations, their cross-talk, and relative influence can help devise single or combinatorial biologic candidates that can tilt the inflammatory balance in a way that can be beneficial in the later or early stages of OA progression.

Our data also served to redefine the cartilage stem and progenitor-like populations that reside in adult cartilage. We validated the existence of CD105/CD90-, NOTCH1-, and STRO1-expressing CPCs that have been previously described in OA and are highly inflammatory. In addition, we described other CPC populations in OA cartilage that express CD90 and CD105 but are low in inflammation. It will be interesting to compare the regenerative potential of these different subpopulations of CPCs, especially in a low-inflammation microenvironment. Since CD24 is a marker for younger chondrocytes with a higher regenerative potential, it is possible that our combinatorial treatment can boost regenerative populations in addition to mitigating inflammation. Our data also reveal that CD24 expression is associated with mitochondrial biogenesis, another characteristic associated with younger healthy chondrocytes. The data also reveal CPC I as progenitor populations that are lost in OA. Future studies are needed to determine how these CPCs are lost during OA progression and whether reintroduction of these CPCs can benefit cartilage regeneration. A particularly interesting subgroup to follow is the CD73-expressing cells, as CD73 has recently been identified to characterize the hSSCs in bone marrow, which can self-renew and give rise to cartilage, bone, and fat progenitor cells (5).

By characterizing chondrocyte populations in patients with OA, we stratified patients by the abundance of each population. This practice is well established in the cancer field, where patient heterogeneity and tumor subtyping play an ever-increasing role in precision medicine. Identification of the 20 different subpopulations in cartilage allowed revealed three major categories of patients with OA. Group A represents 60% of the patients, while groups B and C represent 15% each. Group C patients were distinguished from group A and B patients by an expansion of the inflammatory NOTCH1/STRO1-expressing CPCs, which are also highly active in proinflammatory pathways such as NF-B and HIF2A. Group B patients had an expansion of the Inf-D population. A subset of patients driven by inflammation has been suggested previously as well on the basis of RNA sequencing (38) and DNA methylation patterns (39, 40) in cartilage. Future work will reveal the molecular mechanism(s) that drive this heterogeneity, which may be related to the multifactorial etiology of OA that is affected by the genetic, epigenetic, metabolic, as well as lifestyle factors of the patient populations. Such work will also benefit from studying the interactions of the CPCs and Inf-A and Inf-D cells in multifactorial systems that take into account all the other cell types present in the joint.

In summary, this study provides the first high-dimensional cyTOF map for adult cartilage, revealing multiple, rare subpopulations that coexist in health and disease. Collectively, our data highlight the complex interplay between Inf-A and Inf-D populations and regenerative populations in cartilage and suggest that altering the balance between these populations could provide novel therapeutic strategies for OA. In future studies, refined panels and larger cohort sizes can provide a powerful platform for the stratification of patients with OA based on the underlying cellular drivers of their disease. Ultimately, these stratification efforts would allow for targeted testing of drugs for each patient subset, to establish personalized medicine strategies for OA.

Research objectives. Our objective was to profile rare populations of CPCs in OA patient samples and determine their interactions. We designed a curated panel of antibodies (see below) and tested a cohort of 20 OA patient and 5 normal samples. Observations from this dataset were then more thoroughly tested.

Research subjects. Chondrocytes were derived from OA cartilage or healthy cartilage samples. All experiments were performed on primary cells.

Experimental design. We collected a cohort of 20 patients, which passed several quality control parameters (see below) and included a variety of ages and a balanced pool of male/female patients. Samples that did not pass quality control metrics were not used for downstream analysis. Patient samples were selected on the basis of previously established quality control criteria, namely, the expression ratio of Col2a1/Col1a1 (see methods below) and the expression of MMP genes. Follow-up analysis was conducted on a separate panel of OA chondrocytes to ensure that we could independently see the same results.

Blinding. Researchers were not blind to disease status or treatment when analyzing the data.

Data inclusion/exclusion criteria. All collected data points were used for assays performed after drug treatment. All datasets were quality controlled, and wells or data points that did not pass quality control metrics were not used. This includes (i) Luminex wells that did not give acceptable standard bead readings, (ii) quantitative polymerase chain reaction (qPCR) wells that did not give suitable Ct values for Actin, and (iii) cells analyzed by cyTOF that did not have high SOX9 or CD44 expression. Quality control exclusions were performed before analysis of data. After exclusion of points for these reasons, no additional points were excluded.

Replicates. All drug treatments were performed in independent technical replicates for each patient (i.e., cells derived from the same patient were treated three times with drug versus control). All drug treatments were performed in three to six patient samples.

OA samples were procured from the discarded tissues of patients with radiographic OA undergoing total joint replacement, in accordance with the IRB protocol approved by Stanford University, as previously described (9). The age range for OA patient samples was 54 to 72 years old. Cartilage was shaved from the underlying bone, allowed to recover overnight at 37C in complete media [HyClone Dulbecco's modified Eagle's medium:F12 (GE Healthcare, SH3002302) supplemented with 2 mM l-glutamine (Gibco, 25-030-149), 10% fetal bovine serum (FBS) (Corning, 35-016-CV), 1 antibiotic-antimycotic (Gibco, 15-240-062), and ascorbic acid (12.5 g/ml; Eastman)] and then treated with collagenase (Collagenase II and IV, 2.5 mg/ml each; Worthington Biochem) in complete media overnight at 37C. The next day, cells were strained, centrifuged, and plated at a high density of 2.6 104 cells/cm in complete media. Cells were allowed to become confluent on the plates and were passaged once using collagenase, before cyTOF experiments or drug treatments. Samples were checked for Col2a1/Col1a1 ratios and MMP3, MMP9, and MMP13 expression, before experimentation. Normal samples were either derived from expired cartilage allograft samples or shipped from the manufacturer (samples 1 to 4) or from the surgical waste of a notchplasty (sample 5) under an approved IRB and processed as described above.

Cells for RNA extraction were collected in RNA lysis buffer (Zymo Research) and processed according to the manufacturers specifications for the Quick-RNA MicroPrep Kit (Zymo Research, R1051), including the optional deoxyribonuclease I digestion. RNA quality and quantity were measured using the NanoDrop 1000 Spectrophotometer. All samples had A260/280 (absorbance at 260 and 280 nm) scores between 1.6 and 1.8.

Gene expression analyses. One milligram of RNA from each sample was reversed transcribed into complementary DNA (cDNA) using the High-Capacity cDNA Reverse Transcription Kit (Applied Biosystems, 4368813). qPCR was performed using TaqMan gene-specific expression assays, FAM (carboxyfluoresceinlabeled, for metalloproteinases 3, 9, and 13 (Hs00233962_m1, Hs00957562_m1, and Hs00233992_m1), with a universal master mix (Applied Biosystems, 4369016). Gene expression levels were normalized with FAM-labeled -actin (Hs01060665_g1).

For Tfam, CD24, and PGC1a, we used the SybrGreen mastermix (Applied Biosystems, A25742) according to the manufacturers specifications. Primer sequences were as follows: Tfam_F: 5-GCTCAGAACCCAGATGCA AAA-3, Tfam_R: 5-AGGAAGTTCCCTCCAACGC-3; PGC1a_F: 5-CCATGGATGAAGGGTACTTTTCTG-3, PGC1a_R: 5-CTTTTACCAAAGCAGCAGCC-3; CD24_F: 5-TACCCACGCAGATTTATT-3, CD24_R: 5-AGA GTGAGACCACGAAGA-3; Actin_F: 5-CACCAACTGGGACGACAT-3, Actin_R: 5-ACAGCCTGGATAGCAACG-3. qPCR reactions included a 2-min incubation at 50C to inactivate previous amplicons with uracil-DNA glycosylase, followed by a 10-min incubation at 95C to activate the Taq polymerase. The amplification cycle, consisting of 15 s at 95C and 1 min at 60C, was repeated 40 times. The relative expression levels were determined using the Ct method (Ct gene of interest Ct internal control), and relative gene expression is calculated using 2 Ct method and plotted.

OA cells were seeded at high density in 12-well plates and treated with control [dimethyl sulfoxide (DMSO)] or drug the next day for 48 hours. Drug doses were determined on the basis of prior literature and validation: 0.5 mM IBMX (Sigma-Aldrich, I5879) (31), 50 M JNK inhibitor II (Calbiochem, 420119), 25 M NF-B inhibitor BMS-345541 (Sigma-Aldrich, B9935) (28, 41), and 50 M Alk inhibitor SB 431542 hydrate (Sigma-Aldrich, S4317) (42, 43) were used with appropriate dilution in DMSO.

Multiplex autoantibody assay. Cell culture supernatants were collected and spun down at 10,000g for 10 min at 4C to remove any cells or cell debris and then snap-frozen in liquid nitrogen before performing the assay. This assay was performed in the Human Immune Monitoring Center at Stanford University. Human 62-plex kits were purchased from eBiosciences/Affymetrix and used according to the manufacturers recommendations with modifications as described below. Briefly, beads were added to a 96-well plate and washed in a BioTek ELx405 washer. Undiluted samples were added to the plate containing the mixed antibody-linked beads and incubated at room temperature (RT) for 1 hour, followed by overnight incubation at 4C with shaking. Cold temperature and RT incubation steps were performed on an orbital shaker at 500 to 600 rpm. Following the overnight incubation, plates were washed in a BioTek ELx405 washer, and then, biotinylated detection antibody was added for 75 min at RT with shaking. The plate was washed as above, and streptavidin-phycoerythrin was added. After incubation for 30 min at RT, wash was performed as above, and reading buffer was added to the wells. Each sample was measured in duplicate. Plates were read using a Luminex 200 instrument with a lower bound of 50 beads per sample per cytokine. Custom assay control beads by Radix Biosolutions were added to all wells.

Antibodies were labeled according to the manufacturers specifications using the MAXPAR X8 Polymer labeling kit (Fluidigm). One tube was used per 100 g of antibody. Antibodies were purchased labeling ready, without additives, whenever possible. Antibodies with carrier components such as albumin or glycerol were cleaned with Melon Gel IgG Purification columns (Thermo Fisher Scientific) after buffer exchange with Zeba Desalt Spin Columns (Thermo Fisher Scientific) as per the manufacturers specifications. Final antibody concentration was measured using a NanoDrop 1000 Spectrophotometer, set to IgG (immunoglobulin G) mode, diluted to the highest round value in W buffer with sodium azide, and stored at 4C for later use. The complete list of conjugated antibodies, metal isotope, clone information, and the manufacturer can be found in table S1.

Metal conjugated antibodies were tested in a three-point dilution curve, centered on their recommended or optimized fluorescence-activated cell sorting (FACS) concentration, with a 10-fold increase and decrease from this center value. Signal-to-noise ratio was compared by staining known negative samples, such as 293 T cells. The lowest concentration that had no increase in signal upon a 10-fold increase in concentration was used for the final staining concentration (see table S1).

OA cells were cultured to confluence in 10-cm dishes. On the collection day, cells were stained with 25 M Idu (5-Iodo-2-deoxyuridine) for 15 min at 37C in the cell incubator and then with 0.5 M cisplatin for 5 min at RT. Cells were then lifted with 0.25% trypsin-EDTA (Gibco) for 15 min at 37C. Trypsin was quenched using media containing 10% FBS, and cell were washed three times with phosphate-buffered saline to remove any trace amounts of trypsin. Cells were fixed after straining through a 35 M strainer in 1.6% paraformaldehyde (PFA) for 10 min at RT. Cells were washed four times with cells staining media, counted, and frozen in 1 millioncell aliquots in a small amount of cell staining media at 80C. To stain, cells were thawed on ice and barcoded using the Cell-ID 20-plex Pd Barcoding Kit (Fluidigm) according to the manufacturers specifications. After barcoding, cells were labeled as previously described (8). Briefly, all barcoded samples were combined into one FACS tube and washed 3 times with cell staining media and stained with the cell surface antibodies for 30 min at RT according to the concentrations in table S1. Cells were then washed 2 times with cell staining media and permeabilized with 1 ml of cold methanol added dropwise with continuous gentle vortexing. Cells were incubated for 10 min on ice, with gentle vortexing every 2 to 3 min to avoid cell clumping, then washed in cell staining media, and stained with the intracellular antibodies for 30 min at RT. After 2 times washed with cell staining media, cells were resuspended in 1.6% PFA with Cell-ID Intercalator-Ir (Fluidigm) used at 1:2000. Cells were measured using the cyTOF 2 (Fluidigm) and injected using the supersampler. EQ (Four Element Calibration) beads (Fluidigm) were added just before runtime (1:10 dilution) to normalize signal over runtime.

Normalization over run time was performed using the EU beads using the previously published bead normalized (v0.3) available here: https://github.com/nolanlab/bead-normalization/releases with the default parameters. Samples were then debarcoded using the single-cell debarcoder available here: https://github.com/nolanlab/single-cell-debarcoder using the default parameters. Channel values were arcsine transformed and normalized between the two independent runs using two patients with OA that were loaded in both runs. The tower-independent runs were normalized to each other. Next, we selected for live cells by gating for cisplatin-negative, DNA (Ir195)positive cells. Last, from live cells, we gated for SOX9/CD44 double-positive cells, which were included in the final analysis. On average, 98% of the OA and normal cells were live, and 95 and 64%, respectively, were in the SOX9/CD44 gate. Gating was performed using Cytobank.

Clusters were called using FlowSOM (10). Analysis was performed using Cytobanks online implementation using the standard settings. Clustering was performed using the cell surface receptors, HIF2A and SOD2 (superoxide dismutase 2); no signaling markers were included. The self-organizing map (SOM) was constructed using the 20 OA and 5 normal samples, and then, the same SOM was applied to the treated samples. tSNE projection was also performed using Cytobanks online platform. All results, including FlowSOM clusters and tSNE coordinates, were exported as text files and manipulated for plotting in Python. We compared the results from our FlowSOM clusters to other clustering algorithms, including X-shift (24), and obtained similar numbers of clusters and patterns of expression within each cluster.

Data were visualized using Python and the NumPy (www.numpy.org/), pandas (https://pandas.pydata.org/pandas-docs/stable/), and seaborn (https://seaborn.pydata.org/) packages. Hierarchical clustering of samples or cell populations was performed using the seaborn clustermap function, using a complete-linkage algorithm, also known as the farthest neighbor clustering, in which clusters are decided on the basis of the two most dissimilar points. Complete linkage clustering avoids the chaining phenomenon that can occur with single-linkage methods, where clusters that may be very distant from each other are forced together because of a single element being close. Complete linkage tends to find compact clusters of equal diameters.

Gene counts were downloaded from Gene Expression Omnibus and reanalyzed using custom Python scripts. Gene expression networks and pathway analyses were performed using Ingenuity Pathway Analysis (QIAGEN), Enrichr, and STRING.

Planned comparisons were performed with the GraphPad Prism software. We used (i) one-way analysis of variance (ANOVA) followed by Tukeys post hoc test to identify specific differences between drug treatment groups or between selected OA patient groups (for treatments, groups were only compared against DMSO controls, not against each other) and (ii) nonparametric, two-tailed Welchs t test for comparisons between only two groups. P values were corrected for multiple hypothesis testing, such that the family-wise error was capped at 0.05, using the Bonferroni correction method. The exact method and specific P values for significant comparisons are stated in the appropriate results section. For cyTOF plots, although only 9000 cells were visualized on the tSNE plots in the figures, average values and other calculations or statistics were performed with all cells that met the required criteria.

Acknowledgments: We would like to thank Y. Rosenberg-Hasson at the Stanford Human Immune Profiling Center for help with the Luminex analysis, E. Migliore for help with sample acquisition, and N. Sahu for helpful feedback on the manuscript. Funding: P.S. and N.B. are supported by NIH/NIAMS grants R01 AR070865 and R01 AR070864, F.C.G. is supported by the NSF GRFP award. This work was also supported by a gift to the Department of Orthopedic Surgery from K. Thiery and D. OLeary. Author contributions: F.C.G. conceptualized and designed the experiments, executed the study, analyzed and interpreted the data, and wrote the manuscript. R.B. and S.B. helped with the technical details of acquiring the data. P.S. assisted with data interpretation. S.M. assisted with data collection of the initial IBMX data. S.G., D.F.A., P.F.I., and C.C. provided OA or normal cartilage samples. N.B. conceptualized and designed the study, oversaw data collection, and wrote the manuscript. Competing interests: Patent Methods and Compositions for the Treatment of Osteoarthritis is provisionally filed. The organization issuing the patent is The Board of Trustees of the Leland Stanford Junior University, and the applicants are N.B. and F.C.G. (filed 2 October 2019; serial number 62/909,547). All other 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.

More:
Single-cell mass cytometry reveals cross-talk between inflammation-dampening and inflammation-amplifying cells in osteoarthritic cartilage - Science...

‘I’m aware I could die, but you can’t live in fear’: Living with cancer amid coronavirus – The Age

Ms Chapman's immune system has been severely suppressed since she started treatment for myeloma, a type of blood cancer, 10 years ago.

Coronavirus has spread to at least 200 cases and three deaths in Australia, the country bracing for the full impact of what the World Health Organisation on Friday declared a pandemic.

Whatever protections wary Australians have taken up against coronavirus in the last month, Ms Chapman has been doing since 2010 when she was told she had three to five years to live.

Now, while she isn't letting fear rule her life, her usual precautions are even more critical.

About 110,000 people in Australia currently live with blood cancers or blood-related disorders such as leukemia, myeloma and lymphoma, with 41 new cases diagnosed every day.

Blood cancers in particular suppress the immune system, while chemotherapy and medication for any cancer heighten the danger of viruses.

Ms Chapman, an author, has to pick the events she attends carefully and be aware at all times - but doesn't let fear rule her life.Credit:Luis Enrique Ascui

Myeloma patients never fully recover; while in remission they know the cancer, which weakens your bones and causes extreme fatigue, could return at any time.

"Our patients are always susceptible it's just whether it's high risk or ultra high risk," said Steve Roach, chief executive of Myeloma Australia.

The average age of myeloma onset is 65, placing many patients in a double-risk group.

Ms Chapman, 64, takes a taxi or drives everywhere at the moment. She's avoiding shopping centres and restaurants that are too busy.

She had decided to skip the Melbourne Comedy Festival, which normally brings her great joy, even before it was cancelled. If she goes to the cinema, she'll sit in an aisle seat at the back for a "speedy exit if I hear coughing or spluttering".

Ms Chapman, an author, takes 11 medications every day and has stocked up on supply for the next few weeks - but she still planned to attend a fun run on Sunday, until it was cancelled due to coronavirus fears.

"I'm not pretending these things aren't happening. I'm aware if I catch coronavirus I could be very sick and die from it. But you can't live your life in fear," she said.

"I could stay home, not go out, just go to my appointments. But that's not living either. I'm just being as cautious as I can. That might be really silly, but I'm inclined to just get on with things."

Ms Chapman had stem cell treatment soon after her diagnosis intense chemotherapy that tries to blast as much of the cancer out of your body as possible.

"If that was now, I would be incredibly vulnerable. You actually feel that you're dying, the lethargy is incredible. I would not be going out at all," she said.

Loading

Research on coronavirus' impact on cancer patients is limited, however one study from China found that of 18 coronavirus cases with a history of cancer, almost half had a higher risk of requiring ventilation or of death.

Professor Simon Harrison from the Peter MacCallum Cancer Centre says his current advice to cancer patients, along with good hygiene and avoiding close contact with others in closed spaces, is to assess the risk in every scenario.

"What are the risks? How important is it to you? As with any virus, it needs a community response. We should all ask: if I have an infection, can I stay home for the benefit of others?" Professor Harrison said.

The social impact is unavoidable: Myeloma Australia runs 52 support groups for patients that will now run online for the foreseeable future.

"The challenge for us now is how do we keep the community engaged with each other and feeling supported," Mr Roach said.

"A big thing for us is the bonds that are created in our groups. This coronavirus situation is only going to isolate people even more."

Cancer Council Australia's information and support line is available on 13 11 20.

Michael is a reporter for The Age.

Go here to see the original:
'I'm aware I could die, but you can't live in fear': Living with cancer amid coronavirus - The Age

Global induced pluripotent stem cells market is expected to grow with a CAGR of 8.6% over the forecast period from 2019-2025 – GlobeNewswire

New York, March 13, 2020 (GLOBE NEWSWIRE) -- Reportlinker.com announces the release of the report "Induced Pluripotent Stem Cells Market: Global Industry Analysis, Trends, Market Size, and Forecasts up to 2025" - https://www.reportlinker.com/p05874276/?utm_source=GNW 6% over the forecast period from 2019-2025. The study on induced pluripotent stem cells market covers the analysis of the leading geographies such as North America, Europe, Asia-Pacific, and RoW for the period of 2017 to 2025.

The report on induced pluripotent stem cells market is a comprehensive study and presentation of drivers, restraints, opportunities, demand factors, market size, forecasts, and trends in the global induced pluripotent stem cells market over the period of 2017 to 2025. Moreover, the report is a collective presentation of primary and secondary research findings.

Porters five forces model in the report provides insights into the competitive rivalry, supplier and buyer positions in the market and opportunities for the new entrants in the global induced pluripotent stem cells market over the period of 2017 to 2025. Further, IGR- Growth Matrix gave in the report brings an insight into the investment areas that existing or new market players can consider.

Report Findings1) Drivers Increased government fundings and rising industry focus on the development of novel therapies Rising interest in stem cell therapy2) Restraints High the cost associated with storage3) Opportunities Growing applications of iPS cells in several biopharmaceutical applications provides extensive potential to the key players in the market

Research Methodology

A) Primary ResearchOur primary research involves extensive interviews and analysis of the opinions provided by the primary respondents. The primary research starts with identifying and approaching the primary respondents, the primary respondents are approached include1. Key Opinion Leaders associated with Infinium Global Research2. Internal and External subject matter experts3. Professionals and participants from the industry

Our primary research respondents typically include1. Executives working with leading companies in the market under review2. Product/brand/marketing managers3. CXO level executives4. Regional/zonal/ country managers5. Vice President level executives.

B) Secondary ResearchSecondary research involves extensive exploring through the secondary sources of information available in both the public domain and paid sources. At Infinium Global Research, each research study is based on over 500 hours of secondary research accompanied by primary research. The information obtained through the secondary sources is validated through the crosscheck on various data sources.

The secondary sources of the data typically include1. Company reports and publications2. Government/institutional publications3. Trade and associations journals4. Databases such as WTO, OECD, World Bank, and among others.5. Websites and publications by research agencies

Segment CoveredThe global induced pluripotent stem cells market is segmented on the basis of derived cell type, application, and end user.

The Global Induced Pluripotent Stem Cells Market by Derived Cell Type Fibroblasts Amniotic Cells Hepatocytes Keratinocytes Others

The Global Induced Pluripotent Stem Cells Market by Application Drug Development Regenerative Medicine Toxicity Testing Academic Research

The Global Induced Pluripotent Stem Cells Market by End User Research Organizations Hospitals Biopharma Industries

Company Profiles Astellas Pharma Inc. Fate Therapeutics Inc. FUJIFILM Holdings Corporation Evotec SE Japan Tissue Engineering Co., Ltd ViaCyte, Inc. Vericel Corporation Bristol-Myers Squibb Company Aastrom Biosciences, Inc. Acelity Holdings, Inc.

What does this report deliver?1. Comprehensive analysis of the global as well as regional markets of the induced pluripotent stem cells market.2. Complete coverage of all the segments in the induced pluripotent stem cells market to analyze the trends, developments in the global market and forecast of market size up to 2025.3. Comprehensive analysis of the companies operating in the global induced pluripotent stem cells market. The company profile includes analysis of product portfolio, revenue, SWOT analysis and latest developments of the company.4. IGR- Growth Matrix presents an analysis of the product segments and geographies that market players should focus to invest, consolidate, expand and/or diversify.Read the full report: https://www.reportlinker.com/p05874276/?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.

__________________________

Read the original post:
Global induced pluripotent stem cells market is expected to grow with a CAGR of 8.6% over the forecast period from 2019-2025 - GlobeNewswire

New drugs are costly and unmet need is growing – The Economist

Mar 12th 2020

BEING ABLE to see all the details of the genome at once necessarily makes medicine personal. It can also make it precise. Examining illness molecule by molecule allows pharmaceutical researchers to understand the pathways through which cells act according to the dictates of genes and environment, thus seeing deep into the mechanisms by which diseases cause harm, and finding new workings to target. The flip side of this deeper understanding is that precision brings complexity. This is seen most clearly in cancer. Once, cancers were identified by cell and tissue type. Now they are increasingly distinguished by their specific genotype that reveals which of the panoply of genes that can make a cell cancerous have gone wrong in this one. As drugs targeted against those different mutations have multiplied, so have the options for oncologists to combine them to fit their patients needs.

Cancer treatment has been the most obvious beneficiary of the genomic revolution but other diseases, including many in neurology, are set to benefit, too. Some scientists now think there are five different types of diabetes rather than two. There is an active debate about whether Parkinsons is one disease that varies a lot, or four. Understanding this molecular variation is vital when developing treatments. A drug that works well on one subtype of a disease might fail in a trial that includes patients with another subtype against which it does not work at all.

Thus how a doctor treats a disease depends increasingly on which version of the disease the patient has. The Personalised Medicine Coalition, a non-profit advocacy group, examines new drugs approved in America to see whether they require such insights in order to be used. In 2014, it found that so-called personalised medicines made up 21% of the drugs newly approved for use by Americas Food and Drug Administration (FDA). In 2018 the proportion was twice that.

Two of those cited were particularly interesting: Vitrakvi (larotrectinib), developed by Loxo Oncology, a biotech firm, and Onpattro (patisiran), developed by Alnylam Pharmaceuticals. Vitrakvi is the first to be approved from the start as tumour agnostic: it can be used against any cancer that displays the mutant protein it targets. Onpattro, which is used to treat peripheral-nerve damage, is the first of a new class of drugssmall interfering RNAs, or siRNAsto be approved. Like antisense oligonucleotides (ASOs), siRNAs are little stretches of nucleic acid that stop proteins from being made, though they use a different mechanism.

Again like ASOs, siRNAs allow you to target aspects of a disease that are beyond the reach of customary drugs. Until recently, drugs were either small molecules made with industrial chemistry or bigger ones made with biologynormally with genetically engineered cells. If they had any high level of specificity, it was against the actions of a particular protein, or class of proteins. Like other new techniques, including gene therapies and anti-sense drugs, siRNAs allow the problem to be tackled further upstream, before there is any protein to cause a problem.

Take the drugs that target the liver enzyme PCSK9. This has a role in maintaining levels of bad cholesterol in the blood; it is the protein that was discovered through studies of families in which congenitally high cholesterol levels led to lots of heart attacks. The first generation of such drugs were antibodies that stuck to the enzyme and stopped it working. However, the Medicines Company, a biotech firm recently acquired by Novartis, won approval last year for an siRNA called inclisiran that interferes with the expression of the gene PCSK9thus stopping the pesky protein from being made in the first place. Inclisiran needs to be injected only twice a year, rather than once a month, as antibodies do.

New biological insights, new ways of analysing patients and their disease and new forms of drug are thus opening up a wide range of therapeutic possibilities. Unfortunately, that does not equate to a range of new profitable opportunities.

Thanks in part to ever better diagnosis, there are now 7,000 conditions recognised as rare diseases in America, meaning that the number of potential patients is less than 200,000. More than 90% of these diseases have no approved treatment. These are the diseases that personalised, precision medicine most often goes after. Nearly 60% of the personalised medicines approved by the FDA in 2018 were for rare diseases.

Zolgensma is the most expensive drug ever brought to market.

That might be fine, were the number of diseases stable. But precision in diagnosis is increasingly turning what used to be single diseases into sets of similar-looking ones brought about by distinctly different mechanisms, and thus needing different treatment. And new diseases are still being discovered. Medical progress could, in short, produce more new diseases than new drugs, increasing unmet need.

Some of it will, eventually, be met. For one thing, there are government incentives in America and Europe for the development of drugs for rare diseases. And, especially in America, drugs for rare diseases have long been able to command premium prices. Were this not the case, Novartis would not have paid $8.7bn last year to buy AveXis, a small biotech firm, thereby acquiring Zolgensma, a gene therapy for spinal muscular atrophy (SMA). Most people with SMA lack a working copy of a gene, SMN1, which the nerve cells that control the bodys muscles need to survive. Zolgensma uses an empty virus-like particle that recognises nerve cells to deliver working copies of the gene to where it is needed. Priced at $2.1m per patient, it is the most expensive drug ever brought to market. That dubious accolade might not last long. BioMarin, another biotech firm, is considering charging as much as $3m for a forthcoming gene therapy for haemophilia.

Drug firms say such treatments are economically worthwhile over the lifetime of the patient. Four-fifths of children with the worst form of SMA die before they are four. If, as is hoped, Zolgensma is a lasting cure, then its high cost should be set against a half-century or more of life. About 200 patients had been treated in America by the end of 2019.

But if some treatments for rare diseases may turn a profit, not all will. There are some 6,000 children with SMA in America. There are fewer than ten with Jansens disease. When Dr Nizar asked companies to help develop a treatment for it, she says she was told your disease is not impactful. She wrote down the negative responses to motivate herself: Every day I need to remind myself that this is bullshit.

A world in which markets shrink, drug development gets costlier and new unmet needs are ceaselessly discovered is a long way from the utopian future envisaged by the governments and charities that paid for the sequencing of all those genomes and the establishment of the worlds biobanks. As Peter Bach, director of the Centre for Health Policy and Outcomes, an academic centre in New York, puts it with a degree of understatement: if the world needs to spend as much to develop a drug for 2,000 people as it used to spend developing one for 100,000, the population-level returns from medical research are sharply diminishing.

And it is not as if the costs of drug development have been constant. They have gone up. What Jack Scannell, a consultant and former pharmaceutical analyst at UBS, a bank, has dubbed Erooms lawEroom being Moore, backwardsshows the number of drugs developed for a given amount of R&D spending has fallen inexorably, even as the amount of biological research skyrocketed. Each generation assumes that advances in science will make drugs easier to discover; each generation duly advances science; each generation learns it was wrong.

For evidence, look at the way the arrival of genomics in the 1990s lowered productivity in drug discovery. A paper in Nature Reviews Drug Discovery by Sarah Duggers from Columbia University and colleagues argues that it brought a wealth of new leads that were difficult to prioritise. Spending rose to accommodate this boom; attrition rates for drugs in development subsequently rose because the candidates were not, in general, all that good.

Today, enthused by their big-science experience with the genome and enabled by new tools, biomedical researchers are working on exhaustive studies of all sorts of other omes, including proteomesall the proteins in a cell or body; microbiomesthe non-pathogenic bacteria living in the mouth, gut, skin and such; metabolomessnapshots of all the small molecules being built up and broken down in the body; and connectomes, which list all the links in a nervous system. The patterns they find will doubtless produce new discoveries. But they will not necessarily, in the short term, produce the sort of clear mechanistic understanding which helps create great new drugs. As Dr Scannell puts it: We have treated the diseases with good experimental models. Whats left are diseases where experiments dont replicate people. Data alone canot solve the problem.

Daphne Koller, boss of Insitro, a biotech company based in San Francisco, shares Dr Scannells scepticism about the way drug discovery has been done. A lot of candidate drugs fail, she says, because they aim for targets that are not actually relevant to the biology of the condition involved. Instead researchers make decisions based on accepted rules of thumb, gut instincts or a ridiculous mouse model that has nothing to do with what is actually going on in the relevant human diseaseeven if it makes a mouse look poorly in a similar sort of way.

But she also thinks that is changing. Among the things precision biology has improved over the past five to 10 years have been the scientists own tools. Gene-editing technologies allow genes to be changed in various ways, including letter by letter; single-cell analysis allows the results to be looked at as they unfold. These edited cells may be much more predictive of the effects of drugs than previous surrogates. Organoidsself-organised, three-dimensional tissue cultures grown from human stem cellsoffer simplified but replicable versions of the brain, pancreas, lung and other parts of the body in which to model diseases and their cures.

Insitro is editing changes into stem cellswhich can grow into any other tissueand tracking the tissues they grow into. By measuring differences in the development of very well characterised cells which differ in precisely known ways the company hopes to build more accurate models of disease in living cells. All this work is automated, and carried out on such a large scale that Dr Koller anticipates collecting many petabytes of data before using machine learning to make sense of it. She hopes to create what Dr Scannell complains biology lacks and what drug designers need: predictive models of how genetic changes drive functional changes.

There are also reasons to hope that the new upstream drugsASOs, siRNAs, perhaps even some gene therapiesmight have advantages over todays therapies when it comes to small-batch manufacture. It may also prove possible to streamline much of the testing that such drugs go through. Virus-based gene-therapy vectors and antisense drugs are basically platforms from which to deliver little bits of sequence data. Within some constraints, a platform already approved for carrying one message might be fast-tracked through various safety tests when it carries another.

One more reason for optimism is that drugs developed around a known molecule that marks out a diseasea molecular markerappear to be more successful in trials. The approval process for cancer therapies aimed at the markers of specific mutations is often much shorter now than it used to be. Tagrisso (osimertinib), an incredibly specialised drug, targets a mutation known to occur only in patients already treated for lung cancer with an older drug. Being able to specify the patients who stand to benefit with this degree of accuracy allows trials to be smaller and quicker. Tagrisso was approved less than two years and nine months after the first dose was given to a patient.

With efforts to improve the validity of models of disease and validate drug targets accurately gaining ground, Dr Scannell says he is sympathetic to the proposal that, this time, scientific innovation might improve productivity. Recent years have seen hints that Erooms law is being bent, if not yet broken.

If pharmaceutical companies do not make good on the promise of these new approaches then charities are likely to step in, as they have with various ASO treatments for inherited diseases. And they will not be shackled to business models that see the purpose of medicine as making drugs. The Gates Foundation and Americas National Institutes of Health are investing $200m towards developing treatments based on rewriting genes that could be used to tackle sickle-cell disease and HIVtreatments that have to meet the proviso of being useful in poor-country clinics. Therapies in which cells are taken out of the body, treated in some way and returned might be the basis of a new sort of business, one based around the ability to make small machines that treat individuals by the bedside rather than factories which produce drugs in bulk.

There is room in all this for individuals with vision; there is also room for luck: Dr Nizar has both. Her problem lies in PTH1R, a hormone receptor; her PTH1R gene makes a form of it which is jammed in the on position. This means her cells are constantly doing what they would normally do only if told to by the relevant hormone. A few years ago she learned that a drug which might turn the mutant receptor off (or at least down a bit) had already been characterisedbut had not seemed worth developing.

The rabbit, it is said, outruns the fox because the fox is merely running for its dinner, while the rabbit is running for its life. Dr Nizars incentives outstrip those of drug companies in a similar way. By working with the FDA, the NIH and Massachusetts General Hospital, Dr Nizar helped get a grant to make enough of the drug for toxicology studies. She will take it herself, in the first human trial, in about a years time. After that, if things go well, her childrens pain may finally be eased.

This article appeared in the Technology Quarterly section of the print edition under the headline "Kill or cure?"

Link:
New drugs are costly and unmet need is growing - The Economist

Sit, stay forever: Americans willing to pay top dollar to … – Thomson Reuters Foundation

By Barbara Goldberg

PHILADELPHIA, March 12 (Reuters) - On a follow-up visit after Sophie Cortellino's life-saving cardiac procedure, Dr. Anna Gelzer was delighted and family members were relieved to see her responding so well. Sophie agreed, her tail wagging excitedly.

As the 9-year-old boxer lay on a metal table, Gelzer tried to push up her heart rate as part of a stress test following the procedure in August - a ventricular ablation for an arrhythmia, or irregular heartbeat, the first performed on a dog in the United States.

"You want a cookie?!" Gelzer teased. "Want to go for a walk?!"

The jagged line tracing Sophie's heartbeat on a monitor spiked dramatically but she lay calm and alert, and Gelzer grinned with satisfaction.

Sophie is one of countless aging American dogs undergoing cardiac treatments, stem cell transplants, tracheal stents, pacemakers and other sophisticated, expensive procedures to prolong their lives.

Owners of the dogs, many of whom have been around long enough to watch children grow up and provide support through countless family joys and traumas, are going to great lengths to prolong their lives, paying bills of up to $3,000 for stem cell therapy for arthritis and $7,000 for cardiac procedures like Sophie's.

"Dogs are like a person, a family member," said Gelzer, cardiology professor at the University of Pennsylvania's School of Veterinary Medicine in Philadelphia.

Nearly half of the nation's 77 million pet dogs are aged 6 or older, a 15 percent increase since 1987, according to the American Veterinary Medical Association.

Larger breed dogs are considered geriatric at 6 years old, smaller breeds at 7 years old.

Aging dogs contend with many of the same illnesses as elderly humans, including heart disease, diabetes and senility. Cancer ravages canines at roughly the same rate as humans, striking nearly half of all dogs over age 10, experts say.

In Sophie's case, it was an arrhythmia, a condition that has been treated successfully in humans.

In the procedure, a catheter was snaked through blood vessels into her heart's lower chamber, which pumps oxygen-rich blood to the body, and trouble spots were cauterized. Gelzer was joined in the operation by her counterpart who handles human patients, Dr. Cory Tschabrunn, on the Philadelphia campus.

LOYAL COMPANION

Karen Cortellino, a radiologist, recalled the time nearly a decade ago when she first met Sophie, an 8-month-old puppy who had been rejected by another family. Sophie has since helped celebrate Cortellino's son's college graduation and her daughter's law school commencement, and dressed up as a member of a family wedding party.

When Cortellino's elderly mother moved into their home in Montville, New Jersey, Sophie was her sole companion after others left each morning for work or school.

"They were the best of friends. My mom died last May and Sophie was there for that," Cortellino said. "I would definitely sacrifice what I needed to in order for Sophie to have any procedure that would help her live longer - but well."

When Sophie collapsed last summer and was diagnosed with an arrhythmia, Gelzer had just won a grant for a clinical trial to test canine ventricular ablations, so this procedure was free.

The eventual cost is likely to be $5,000-$7,000, Gelzer said, which Cortellino said she would be "absolutely" willing to pay if Sophie needs another one.

Her exam completed, Gelzer gladly accepted sloppy kisses from her patient.

"There is never a doubt that what we do is meaningful," Gelzer said. "You have to enjoy working with the animals and with the owners. I like both - very much." (Reporting by Barbara Goldberg Editing by Sonya Hepinstall)

Our Standards: The Thomson Reuters Trust Principles.

See the original post here:
Sit, stay forever: Americans willing to pay top dollar to ... - Thomson Reuters Foundation

Second patient cured of HIV using stem cell transplant treatment – The Japan Times

PARIS A second patient has been cured of HIV after undergoing stem cell transplant treatment, doctors said Tuesday, after finding no trace of infection 30 months after he stopped traditional treatment.

The London Patient, a cancer sufferer originally from Venezuela, made headlines last year when researchers at the University of Cambridge reported they had found no trace of the AIDS-causing virus in his blood for 18 months.

Ravindra Gupta, lead author of the study published in The Lancet HIV, said the new test results were even more remarkable and likely demonstrated the patient was cured.

Weve tested a sizeable set of sites that HIV likes to hide in and they are all pretty much negative for an active virus, Gupta told AFP.

The patient, who revealed his identity this week as Adam Castillejo, 40, was diagnosed with HIV in 2003 and had been on medication to keep the disease in check since 2012.

Later that year, he was diagnosed with advanced Hodgkins lymphoma, a deadly cancer.

In 2016 he underwent a bone marrow transplant to treat blood cancer, receiving stem cells from donors with a genetic mutation present in less than 1 percent of Europeans that prevents HIV from taking hold.

He becomes only the second person to be cured of HIV after American Timothy Brown, known as the Berlin Patient, recovered from HIV in 2011 following similar treatment.

Viral tests of Castillejos cerebral fluid, intestinal tissue and lymphoid tissue more than two years after stopping retroviral treatment showed no active infection.

Gupta said the tests uncovered HIV fossils fragments of the virus that were now incapable of reproducing, and were therefore safe.

Wed expect that, he said.

Its quite hard to imagine that all trace of a virus that infects billions of cells was eliminated from the body.

Researchers cautioned that the breakthrough did not constitute a generalized cure for HIV, which leads to nearly 1 million deaths every year.

Castillejos treatment was a last resort as his blood cancer would likely have killed him without intervention, according to Gupta.

The Cambridge doctor said that there were several other patients who had undergone similar treatment but who were less far along in their remission.

There will probably be more but they will take time, he said.

Researchers are currently weighing up whether or not patients suffering from drug-resistant forms of HIV might be eligible for stem cell transplants in future, something Gupta said would require careful ethical consideration.

Youd have to weigh up the fact that theres a 10 percent mortality rate from doing a stem-cell transplant against what the risk of death would be if we did nothing, he said.

Commenting on The Lancet study, Sharon Lewin, an infectious disease expert at the University of Melbourne, said the findings could provide comfort to patients.

But she advised caution.

Given the large number of cells sampled here and the absence of any intact virus, is the London patient cured? she said.

Unfortunately in the end, only time will tell.

Continue reading here:
Second patient cured of HIV using stem cell transplant treatment - The Japan Times

What Jason Hope Says About New Longevity Research – HealthTechZone

Throughout the past decade, various topics related to stem cells have made headlines across all platforms. From being hailed as the most innovative method for eradicating specific diseases, to being protested by various groups and organizations, the use of stem cells has gained national attention repeatedly. With promising initial scientific findings, and avid researchers aiming to solidify the presence of stem cell usage in the realm of science on a normalized basis, increasing numbers of startups, biotech giants, and independent companies are forging ahead with stem cell-related projects. As global connectivity, technological advancements, and the marriage between medicine and technology continues to evolve swiftly, Jason Hope sees stem cells will undoubtedly remaining in the spotlight.

Over 20 years ago, scientists successfully extracted the first human embryonic stem cells, and effectively grew these cells in a lab setting. The remarkable feat of being able to successfully grow the parent cells, which essentially allow for the growth of new cells in the body, was a hopeful moment for the medical sector involved in creating effective regenerative treatments for conditions like heart disease, Alzheimers, stroke, and Parkinsons Disease. Using basic reasoning, the successful regeneration of parent cells could provide the regeneration of undesired cells, leading to anti-aging results, or effective care for many age-related conditions that deteriorate the body over time.

Though this initial breakthrough was promising, the scientific community has not yet made significant strides in bringing stem cell therapy to market in a way that is well-researched, backed by medical associations, and commonly accepted by the scientific community. In fact, the only readily utilized stem cell treatments are related to successfully growing blood cells from matching donors for patients with various blood disorders. According to entrepreneur, philanthropist, and expert in the realm of anti-aging and longevity, Jason Hope, these initial utilization of stem cells are commendable, but require a lot more research in order to maximize the potential widespread benefits of stem cells in medicine.

Hope, who has devoted much of his philanthropic endeavors within the medical industry via groups like the SENS Organization, recognizes that most stem cell implementations are rightfully considered experimental until appropriate research, testing, and development can occur. As an expert in the realm of anti-aging, and the championing of increasing health throughout a lifetime, Jason Hope recognizes the potential distrust that can be formulated by the general public as a result of eager companies making lofty claims or promoting potentially faulty treatments not yet fully vetted by the medical community. Thus, while he remains avidly enthralled by the potential maximization of stem cell therapies, hope supports the long-term research needed to safely, successfully, and effectively generate breakthrough stem cell treatments.

Providing continued backing for the extensive research completed at the SENS (Strategies for Engineered Negligible Senescence) Organization, Hopes contributions aid in the research aiming to create preventative treatments for degenerative diseases and utilizing breakthrough science to increase the overall long-term quality of life for individuals. Instead of focusing on the treatment of symptoms and the disease throughout the progression of the condition, the scientists at SENS work to examine ways to successfully prevent the disease from happening. Through this boundary-pushing work, a lot of their research focuses on stem cell intervention. According to Hope, stem cell treatments for Parkinsons Disease are now in the second stage of clinical trials at SENS. While the process of undergoing such extensive trials may appear slow, it is crucial to maintaining overall public support via successful treatment launches and promising in terms of the long-term possibilities linked to stem cell treatments.

In addition to the research being conducted by SENS, preliminary medical studies are being conducted with a myriad of uses for stem cells. Experimental stem cell transplants of retinal cells were recently utilized in a small research study of macular generation, providing initially promising results for the handful of patients who have received artificially generated retinal cells. Elsewhere, scientists have begun to explore ways to minimize potential rejection of stem cells in organs like the liver, through maximizing the most conducive environment for stem cells to thrive. While these slow-moving vehicles of change are less prominent than startups promising the proverbial Fountain of Youth via experimental stem cell treatments, these medically sound research studies are forming the backbone of stem cell treatment for the future.

As with all scientific and medical innovations, Hope also recognizes the potential risks, hurdles, and roadblocks within the growing field of stem cell research, and integration into medicine. From supply chain concerns to potential long-term side effects, and the risk of overly eager startups making too-lofty claims, Hope understands that the road to the everyday utilization of stem cells remains lengthy and potentially bumpy. However, the proverbial juice may very well be worth the squeeze in this example. As stem cells harvest the potential power to overturn the degenerative effects of some of the most prominent diseases, allow individuals to maintain active health for elongated periods of time, and increase the quality of life for countless individuals, expanding upon the initial promising research is potentially a pivotal point for the medical community and humankind. Though the road to successful scientific integration of stem cells is long, the potential healthcare benefits are limitless, and according to industry experts like Jason Hope, worth investing in, exploring, and championing.

About Jason Hope

An avid entrepreneur, investor, and philanthropist, Jason Hope is a futurist involved in the championing of technological advancement, community involvement, and innovative medical interventions. Deeply passionate about the anti-aging, longevity, and human advancement niche of biomedicine, Hope remains actively involved in various scientific organizations.

After receiving a degree in Finance from ASU, and a subsequent MBA from ASUs W.P. Carey School of Business, Hope developed a successful mobile communications company. Professionally, he currently focuses on investing in startups and developing grant programs for small businesses.

Follow Jason Hope:

LinkedIn | Facebook | Twitter | Medium

See the article here:
What Jason Hope Says About New Longevity Research - HealthTechZone

Pipeline: Investigational Therapies for COVID-19 – Monthly Prescribing Reference

While no specific treatment for coronavirus disease 2019 (COVID-19) is currently available, several therapies are being investigated globally.*

Antivirals

AbbVie: the Company is collaborating with select health authorities and institutions to determine the antiviral activity of lopinavir/ritonavir (Kaletra) against COVID-19.

AIM ImmunoTech: developing Ampligen, a broad-spectrum antiviral that will be tested as a potential treatment for COVID-19 in Japan. A significant survival effect was observed in a trial evaluating mice infected with the earlier Severe Acute Respiratory Syndrome (SARS) coronavirus.

Gilead: developing remdesivir, a broad-spectrum antiviral agent that is being investigated in a double-blinded, placebo-controlled study sponsored by the National Institute of Allergy and Infectious Diseases, part of the National Institutes of Health (NIH). In addition, Gilead is initiating two phase 3 trials to evaluate the safety and efficacy of remdesivir in adults diagnosed with COVID-19, following a rapid review and acceptance by the Food and Drug Administration (FDA) of the investigational new drug filing for the novel antiviral.

Immunotherapies and Other Investigational Therapies

Algernon Pharmaceuticals: developing ifenprodil, an N-methyl-d-aspartate (NDMA) receptor glutamate receptor antagonist, which is being prepared for US clinical trials for COVID-19 based on results of an animal study that showed the investigational therapy significantly reduced acute lung injury and improved survivability in H5N1 infected mice.

CEL-SCI: developing an immunotherapy using LEAPS, a patented T cell modulation peptide epitope delivery technology, to stimulate protective cell-mediated T cell responses and reduce viral load.

Innovation Pharmaceuticals: developing brilacidin, a defensin-mimetic, that mimics the human innate immune system and causes disruption of the membrane of pathogens, leading to cell death. It has already been tested in humans in phase 2 trials for other indications.

Mesoblast Limited: investigating remestemcel-L, an allogeneic mesenchymal stem cell (MSC) product candidate, as a treatment for patients with acute respiratory distress syndrome caused by COVID-19. Remestemcel-L, which is comprised of culture-expanded MSCs derived from the bone marrow of an unrelated donor, is administered in a series of intravenous infusions and is believed to have immunomodulatory properties to counteract inflammatory processes.

Q BioMed: partnering with Mannin Research to develop a potential treatment that addresses vascular leakage and endothelial dysfunction, which may potentially help patients with severe cases of COVID-19.

Takeda: developing an anti-SARS-CoV-2 polyclonal hyperimmune globulin (H-IG) to treat high-risk individuals with COVID-19 (TAK-888). Pathogen-specific antibodies from plasma will be collected from recovered patients (or vaccinated donors in the future) and will be transferred to sick patients to improve the immune response to the infection and increase the chance of recovery.

Tiziana: developing TZLS-501, which has been shown to rapidly deplete circulating levels of interleukin-6 (IL-6) in the blood, a key driver of chronic inflammation. Excessive production of IL-6 is believed to be associated with severe lung damage observed with COVID-19 infections.

Vaccines

Altimmune Inc: developing a single-dose, intranasal vaccine against COVID-19 using its proprietary NasoVAX technology. The vaccine is moving toward animal testing.

Applied DNA Sciences: collaborating with Takis Biotech to develop a DNA vaccine candidate using PCR-based DNA (LinearDNA) manufacturing systems; preclinical testing in animals are expected to begin in the second quarter of 2020.

Codagenix Inc: co-developing a live-attenuated vaccine with the Serum Institute of India using viral deoptimization.

GlaxoSmithKline: collaborating with Clover Biopharmaceuticals to develop a protein-based coronavirus vaccine candidate (COVID-19 S-Trimer) using Clovers proprietary technology (Timer-Tag) and combining it with GSKs pandemic adjuvant system.

Inovio Pharmaceuticals: developing a DNA vaccine (INO-4800) to address COVID-19; human trials to begin in the US in April.

Johnson & Johnson: partnering with the Biomedical Advanced Research and Development Authority (BARDA) to develop a vaccine using Janssens AdVac and PER.C6 technology, which provide the ability to rapidly upscale production of an optimal vaccine candidate.

Moderna Inc: vials of the Companys mRNA vaccine (mRNA-1273) have been shipped to the National Institute of Allergy and Infectious Diseases to be used in a phase 1 study in the US.

Novavax: currently evaluating multiple recombinant nanoparticle vaccine candidates in animal models; initiation of phase 1 testing is expected in late spring of 2020. The COVID-19 vaccine candidates will likely include the saponin-based Matrix-M adjuvant to enhance immune responses.

Sanofi: collaborating with BARDA to develop a vaccine using Sanofis recombinant DNA platform. The DNA sequence encoding the antigen will be combined into the DNA of the baculovirus expression platform and used to produce large quantities of the coronavirus antigen which will be formulated to stimulate the immune system to protect against the virus.

*This list is not all inclusive. Updates will be made as more information becomes available.

Read the original here:
Pipeline: Investigational Therapies for COVID-19 - Monthly Prescribing Reference

Here’s how to treat coronavirus, according to research – Daily Nation

By XINHUAMore by this Author

China has released the seventh version of the diagnosis and treatment guideline on the novel coronavirus disease (Covid-19).

Here are some drugs and therapies that have been recommended by the guideline, and some medicines that have been found to have the potential to defeat the virus and have entered clinical trials.

Chloroquine Phosphate, a widely used anti-malaria and autoimmune disease drug, has been used for more than 70 years.

The drug has been used in treating 285 critically ill Covid-19 patients in a hospital in Wuhan, and no obvious adverse reactions have been found so far.

In the latest version of the treatment guideline, Chloroquine Phosphate is recommended for Covid-19 patients from 18 to 65.

The amount for patients over 50 kg is 500 mg per dose twice a day for seven days.

The guideline also noted that patients should take less than three antiviral drugs.

Tocilizumab, with the common brand name Actemra, is an injectable synthetic protein that blocks the effects of IL-6 in patients with rheumatoid arthritis.

IL-6 is a protein that the body produces when there is inflammation.The latest version of the guideline suggests the use of Tocilizumab in patients with an increasing level of IL-6 and with extensive lesions in both lungs or severe symptoms.

Chinese researchers have found that a cause of death for severe and critically ill patients infected with the novel coronavirus is cytokine storm, an overreaction of the immune system.

These patients are found with a higher level of IL-6 in their blood.

Last month, the increasing level of IL-6 was recommended as a warning sign that the patient's situation could possibly deteriorate.

Currently, the drug is under clinical trials in 14 hospitals in Wuhan and a total of 272 severe patients had been treated with Tocilizumab as of March 5.

Convalescent plasma, processed from the plasma collected from recovered Covid-19 patients, contains a large number of protective antibodies.

As of February 28, 245 Covid-19 patients have received the therapy and 91 cases have shown improvement in clinical indicators and symptoms.

According to health authorities, plasma therapy has proved safe and effective.

4. TRADITIONAL CHINESE MEDICINE

Traditional Chinese Medicine (TCM) has been proven effective in treating Covid-19 patients.

With TCM treatment, patients with mild symptoms have seen their fever or cough alleviated, according to medical experts.

For severely ill patients, TCM helped relieve symptoms and restore blood oxygen saturation, preventing the patients' conditions from developing into critically ill cases.

TCM decoction Qingfei Paidu Soup has been recommended to medical institutions nationwide on February 6 after data analysis on 214 cases.

As of February 29, the decoction is used in 66 designated hospitals in 10 provincial-level regions in China.

Favipiravir, an influenza drug available on overseas markets, has been put in a parallel controlled study in Shenzhen, Guangdong Province, with 80 patients enlisted.

The initial outcome of the trial shows the drug has relatively obvious efficacy and low adverse reactions.

Experts have suggested expanding the trial to further observe and study its effect.

Remdesivir, developed against Ebola infections by American pharmaceutical company Gilead Sciences, has shown fairly good antiviral activity against the novel coronavirus at the cellular level.

Cao Bin, a respiratory expert who is leading the Remdesivir programme, said on Wednesday that two trials for Remdesivir are going on smoothly and China will share the data with the international community after the programme is completed.

Clinical studies on stem cell therapy, which can inhibit the overreaction of the body's immune system, have also been carried out to treat severe patients.

As of February 21, four patients who have received the therapy have been discharged from hospital, and the trial is expected to be further expanded. Currently, three kinds of stem cells mesenchymal, lung and embryonic stem cells are used in treatments.

Researchers usually inject stem cell products into the lungs.

Meanwhile, the Chinese Academy of Sciences has developed a new stem cell drug, CAStem, which has shown promising results in animal experiments.

The research team has applied for urgent assessment by the National Medical Products Administration.

Approvals by the ethics committee, and clinical observation and evaluation, are in progress.

Several research and trials on applying stem cells to treat Covid-19 patients have been carried out in the country.

Continue reading here:
Here's how to treat coronavirus, according to research - Daily Nation