Category Archives: Somatic Stem Cells

Somatic evolution in cancer – Wikipedia

Somatic evolution is the accumulation of mutations and epimutations in somatic cells (the cells of a body, as opposed to germ plasm and stem cells) during a lifetime, and the effects of those mutations and epimutations on the fitness of those cells. This evolutionary process has first been shown by the studies of Bert Vogelstein in colon cancer. Somatic evolution is important in the process of aging as well as the development of some diseases, including cancer.

Cells in pre-malignant and malignant neoplasms (tumors) evolve by natural selection.[1][2] This accounts for how cancer develops from normal tissue and why it has been difficult to cure. There are three necessary and sufficient conditions for natural selection, all of which are met in a neoplasm:

Cells in neoplasms compete for resources, such as oxygen and glucose, as well as space. Thus, a cell that acquires a mutation that increases its fitness will generate more daughter cells than competitor cells that lack that mutation. In this way, a population of mutant cells, called a clone, can expand in the neoplasm. Clonal expansion is the signature of natural selection in cancer.

Cancer therapies act as a form of artificial selection, killing sensitive cancer cells, but leaving behind resistant cells. Often the tumor will regrow from those resistant cells, the patient will relapse, and the therapy that had been previously used will no longer kill the cancer cells. This selection for resistance is similar to the repeatedly spraying crops with a pesticide and selecting for resistant pests until the pesticide is no longer effective.

Modern descriptions of biological evolution will typically elaborate on major contributing factors to evolution such as the formation of local micro-environments, mutational robustness, molecular degeneracy, and cryptic genetic variation.[4] Many of these contributing factors in evolution have been isolated and described for cancer.[5]

Cancer is a classic example of what evolutionary biologists call multilevel selection: at the level of the organism, cancer is usually fatal so there is selection for genes and the organization of tissues[6][7] that suppress cancer. At the level of the cell, there is selection for increased cell proliferation and survival, such that a mutant cell that acquires one of the hallmarks of cancer[3] (see below), will have a competitive advantage over cells that have not acquired the hallmark. Thus, at the level of the cell there is selection for cancer.

The earliest ideas about neoplastic evolution come from Boveri[8] who proposed that tumors originated in chromosomal abnormalities passed on to daughter cells. In the decades that followed, cancer was recognized as having a clonal origin associated with chromosomal aberrations.[9][10][11][12]

Early mathematical modeling of cancer, by Armitage and Doll, set the stage for the future development of the somatic evolutionary theory of cancer. Armitage and Doll explained the cancer incidence data, as a function of age, as a process of the sequential accumulation of somatic mutations (or other rate limiting steps).[13]

Advances in cytogenetics facilitated discovery of chromosome abnormalities in neoplasms, including the Philadelphia chromosome in chronic myelogenous leukemia[14] and translocations in acute myeloblastic leukemia.[15] Sequences of karyotypes replacing one another in a tumor were observed as it progressed.[16][17][18] Researchers hypothesized that cancer evolves in a sequence of chromosomal mutations and selection[6][17][19][20] and that therapy may further select clones.[12]

In 1971, Knudson published the 2-hit hypothesis for mutation and cancer based on statistical analysis of inherited and sporadic cases of retinoblastoma.[21] He postulated that retinoblastoma developed as a consequence of two mutations; one of which could be inherited or somatic followed by a second somatic mutation. Cytogenetic studies localized the region to the long arm of chromosome 13, and molecular genetic studies demonstrated that tumorigenesis was associated with chromosomal mechanisms, such as mitotic recombination or non-disjunction, that could lead to homozygosity of the mutation.[22] The retinoblastoma gene was the first tumor suppressor gene to be cloned in 1986.

Cairns hypothesized a different, but complementary, mechanism of tumor suppression in 1975 based on tissue architecture to protect against selection of variant somatic cells with increased fitness in proliferating epithelial populations, such as the intestine and other epithelial organs.[6] He postulated that this could be accomplished by restricting the number of stem cells for example at the base of intestinal crypts and restraining the opportunities for competition between cells by shedding differentiated intestinal cells into the gut. The essential predictions of this model have been confirmed although mutations in some tumor suppressor genes, including CDKN2A (p16), predispose to clonal expansions that encompass large numbers of crypts in some conditions such as Barrett's esophagus. He also postulated an immortal DNA strand that is discussed at Immortal DNA strand hypothesis.

Nowell synthesized the evolutionary view of cancer in 1976 as a process of genetic instability and natural selection.[1] Most of the alterations that occur are deleterious for the cell, and those clones will tend to go extinct, but occasional selectively advantageous mutations arise that lead to clonal expansions. This theory predicts a unique genetic composition in each neoplasm due to the random process of mutations, genetic polymorphisms in the human population, and differences in the selection pressures of the neoplasm's microenvironment. Interventions are predicted to have varying results in different patients. What is more important, the theory predicts the emergence of resistant clones under the selective pressures of therapy. Since 1976, researchers have identified clonal expansions[23][24][25][26][27][28] and genetic heterogeneity[29][30][31][32][33][34] within many different types of neoplasms.

There are multiple levels of genetic heterogeneity associated with cancer, including single nucleotide polymorphism (SNP),[35] sequence mutations,[30] Microsatellite shifts[29] and instability,[36] loss of heterozygosity (LOH),[34] Copy number variation (detected both by comparative genomic hybridization (CGH),[31] and array CGH,[37]) and karyotypic variations including chromosome structural aberrations and aneuploidy.[32][33][38][39][40] Studies of this issue have focused mainly at the gene mutation level, as copy number variation, LOH and specific chromosomal translocations are explained in the context of gene mutation. It is thus necessary to integrate multiple levels of genetic variation in the context of complex system and multilevel selection.

System instability is a major contributing factor for genetic heterogeneity.[41] For the majority of cancers, genome instability is reflected in a large frequency of mutations in the whole genome DNA sequence (not just the protein coding regions that are only 1.5% of the genome[42]). In whole genome sequencing of different types of cancers, large numbers of mutations were found in two breast cancers (about 20,000 point mutations[43]), 25 melanomas (9,000 to 333,000 point mutations[44]) and a lung cancer (50,000 point mutations and 54,000 small additions and deletions[45]). Genome instability is also referred to as an enabling characteristic for achieving endpoints of cancer evolution.[3]

Many of the somatic evolutionary studies have traditionally been focused on clonal expansion, as recurrent types of changes can be traced to illustrate the evolutionary path based on available methods. Recent studies from both direct DNA sequencing and karyotype analysis illustrate the importance of the high level of heterogeneity in somatic evolution. For the formation of solid tumors, there is an involvement of multiple cycles of clonal and non-clonal expansion.[39][46] Even at the typical clonal expansion phase, there are significant levels of heterogeneity within the cell population, however, most are under-detected when mixed populations of cells are used for molecular analysis. In solid tumors, a majority of gene mutations are not recurrent types,[47] and neither are the karyotypes.[39][41] These analyses offer an explanation for the findings that there are no common mutations shared by most cancers.[48]

The state of a cell may be changed epigenetically, in addition to genetic alterations. The best-understood epigenetic alterations in tumors are the silencing or expression of genes by changes in the methylation of CG pairs of nucleotides in the promoter regions of the genes. These methylation patterns are copied to the new chromosomes when cells replicate their genomes and so methylation alterations are heritable and subject to natural selection. Methylation changes are thought to occur more frequently than mutations in the DNA, and so may account for many of the changes during neoplastic progression (the process by which normal tissue becomes cancerous), in particular in the early stages. For instance, when loss of expression of the DNA repair protein MGMT occurs in a colon cancer, it is caused by a mutation only about 4% of the time, while in most cases the loss is due to methylation of its promoter region.[49] Similarly, when loss of expression of the DNA repair protein PMS2 occurs in colon cancer, it is caused by a mutation about 5% of the time, while in most cases loss of expression is due to methylation of the promoter of its pairing partner MLH1 (PMS2 is unstable in the absence of MLH1).[50] Epigenetic changes in progression interact with genetic changes. For example, epigenetic silencing of genes responsible for the repair of mispairs or damages in the DNA (e.g. MLH1 or MSH2) results in an increase of genetic mutations.

Deficiency of DNA repair proteins PMS2, MLH1, MSH2, MSH3, MSH6 or BRCA2 can cause up to 100-fold increases in mutation frequency[51][52][53] Epigenetic deficiencies in DNA repair gene protein expression have been found in many cancers, though not all deficiencies have been evaluated in all cancers. Epigeneticically deficient DNA repair proteins include BRCA1, WRN, MGMT, MLH1, MSH2, ERCC1, PMS2, XPF, P53, PCNA and OGG1, and these are found to be deficient at frequencies of 13% to 100% in different cancers.[citation needed] (Also see Frequencies of epimutations in DNA repair genes.)

In addition to well studied epigenetic promoter methylation, more recently there have been substantial findings of epigenetic alterations in cancer due to changes in histone and chromatin architecture and alterations in the expression of microRNAs (microRNAs either cause degradation of messenger RNAs or block their translation)[54] For instance, hypomethylation of the promoter for microRNA miR-155 increases expression of miR-155, and this increased miR-155 targets DNA repair genes MLH1, MSH2 and MSH6, causing each of them to have reduced expression.[55]

In cancers, loss of expression of genes occurs about 10 times more frequently by transcription silencing (caused by somatically heritable promoter hypermethylation of CpG islands) than by mutations. As Vogelstein et al. point out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations.[56] In contrast, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 somatically heritable heavily methylated CpG islands in promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa.[57][58][59]

Methylation of the cytosine of CpG dinucleotides is a somatically heritable and conserved regulatory mark that is generally associated with transcriptional repression. CpG islands keep their overall un-methylated state (or methylated state) extremely stably through multiple cell generations.[60]

One common feature of neoplastic progression is the expansion of a clone with a genetic or epigenetic alteration. This may be a matter of chance, but is more likely due to the expanding clone having a competitive advantage (either a reproductive or survival advantage) over other cells in the tissue. Since clones often have many genetic and epigenetic alterations in their genomes, it is often not clear which of those alterations cause a reproductive or survival advantage and which other alterations are simply hitchhikers or passenger mutations (see Glossary below) on the clonal expansion.

Clonal expansions are most often associated with the loss of the p53 (TP53) or p16 (CDKN2A/INK4a) tumor suppressor genes. In lung cancer, a clone with a p53 mutation was observed to have spread over the surface of one entire lung and into the other lung.[27] In bladder cancer, clones with loss of p16 were observed to have spread over the entire surface of the bladder.[61][62] Likewise, large expansions of clones with loss of p16 have been observed in the oral cavity[24] and in Barrett's esophagus.[25] Clonal expansions associated with inactivation of p53 have also appear in skin,[23][63] Barrett's esophagus,[25] brain,[64] and kidney.[65] Further clonal expansions have been observed in the stomach,[66] bladder,[67] colon,[68] lung,[69] hematopoietic (blood) cells,[70] and prostate.[71]

These clonal expansions are important for at least two reasons. First, they generate a large target population of mutant cells and so increase the probability that the multiple mutations necessary to cause cancer will be acquired within that clone. Second, in at least one case, the size of the clone with loss of p53 has been associated with an increased risk of a pre-malignant tumor becoming cancerous.[72] It is thought that the process of developing cancer involves successive waves of clonal expansions within the tumor.[73]

The term "field cancerization" was first used in 1953 to describe an area or "field" of epithelium that has been preconditioned by (at that time) largely unknown processes so as to predispose it towards development of cancer.[74] Since then, the terms "field cancerization" and "field defect" have been used to describe pre-malignant tissue in which new cancers are likely to arise. Field defects, for example, have been identified in most of the major areas subject to tumorigenesis in the gastrointestinal (GI) tract.[75] Cancers of the GI tract that are shown to be due, to some extent, to field defects include head and neck squamous cell carcinoma (HNSCC), oropharyngeal/laryngeal cancer, esophageal adenocarcinoma and esophageal squamous-cell carcinoma, gastric cancer, bile duct cancer, pancreatic cancer, small intestine cancer and colon cancer.

In the colon, a field defect probably arises by natural selection of a mutant or epigenetically altered cell among the stem cells at the base of one of the intestinal crypts on the inside surface of the colon. A mutant or epigenetically altered stem cell, if it has a selective advantage, could replace the other nearby stem cells by natural selection. This can cause a patch of abnormal tissue, or field defect. The figure in this section includes a photo of a freshly resected and lengthwise-opened segment of the colon that may represent a large field defect in which there is a colon cancer and four polyps. The four polyps, in addition to the cancer, may represent sub-clones with proliferative advantages.

The sequence of events giving rise to this possible field defect are indicated below the photo. The schematic diagram shows a large area in yellow indicating a large patch of mutant or epigenetically altered cells that formed by clonal expansion of an initial cell based on a selective advantage. Within this first large patch, a second such mutation or epigenetic alteration may have occurred so that a given stem cell acquired an additional selective advantage compared to the other stem cells within the patch, and this altered stem cell expanded clonally forming a secondary patch, or sub-clone, within the original patch. This is indicated in the diagram by four smaller patches of different colors within the large yellow original area. Within these new patches (sub-clones), the process may have been repeated multiple times, indicated by the still smaller patches within the four secondary patches (with still different colors in the diagram) which clonally expanded, until a stem cell arose that generated either small polyps (which may be benign neoplasms) or else a malignant neoplasm (cancer). These neoplasms are also indicated, in the diagram below the photo, by 4 small tan circles (polyps) and a larger red area (cancer). The cancer in the photo occurred in the cecal area of the colon, where the colon joins the small intestine (labeled) and where the appendix occurs (labeled). The fat in the photo is external to the outer wall of the colon. In the segment of colon shown here, the colon was cut open lengthwise to expose the inner surface of the colon and to display the cancer and polyps occurring within the inner epithelial lining of the colon.

Phylogenetics may be applied to cells in tumors to reveal the evolutionary relationships between cells, just as it is used to reveal evolutionary relationships between organisms and species. Shibata, Tavare and colleagues have exploited this to estimate the time between the initiation of a tumor and its detection in the clinic.[29] Louhelainen et al. have used parsimony to reconstruct the relationships between biopsy samples based on loss of heterozygosity.[76] Phylogenetic trees should not be confused with oncogenetic trees,[77] which represent the common sequences of genetic events during neoplastic progression and do not represent the relationships of common ancestry that are essential to a phylogeny. For an up-to-date review in this field, see Bast 2012.[78]

An adaptive landscape is a hypothetical topological landscape upon which evolution is envisioned to take place. It is similar to Wright's fitness landscape[79][80] in which the location of each point represents the genotype of an organism and the altitude represents the fitness of that organism in the current environment. However, unlike Wright's rigid landscape, the adaptive landscape is pliable. It readily changes shape with changes in population densities and survival/reproductive strategies used within and among the various species.

Wright's shifting balance theory of evolution combines genetic drift (random sampling error in the transmission of genes) and natural selection to explain how multiple peaks on a fitness landscape could be occupied or how a population can achieve a higher peak on this landscape. This theory, based on the assumption of density-dependent selection as the principal forms of selection, results in a fitness landscape that is relatively rigid. A rigid landscape is one that does not change in response to even large changes in the position and composition of strategies along the landscape.

In contrast to the fitness landscape, the adaptive landscape is constructed assuming that both density and frequency-dependent selection is involved (selection is frequency-dependant when the fitness of a species depends not only on that species strategy but also on the strategy of all other species). As such, the shape of the adaptive landscape can change drastically in response to even small changes in strategies and densities.[81]

The flexibility of adaptive landscapes provide several ways for natural selection to cross valleys and occupy multiple peaks without having to make large changes in their strategies. Within the context of differential or difference equation models for population dynamics, an adaptive landscape may actually be constructed using a fitness generating function.[82] If a given species is able to evolve, it will, over time, "climb" the adaptive landscape toward a fitness peak through gradual changes in its mean phenotype according to a strategy dynamic that involves the slope of the adaptive landscape. Because the adaptive landscape is not rigid and can change shape during the evolutionary process, it is possible that a species may be driven to maximum, minimum, or saddle point on the adaptive landscape. A population at a global maximum on the adaptive landscape corresponds an evolutionarily stable strategy (ESS) and will become dominant, driving all others toward extinction. Populations at a minimum or saddle point are not resistant to invasion, so that the introduction of a slightly different mutant strain may continue the evolutionary process toward unoccupied local maxima.

The adaptive landscape provides a useful tool for studying somatic evolution as it can describe the process of how a mutant cell evolves from a small tumor to an invasive cancer. Understanding this process in terms of the adaptive landscape may lead to the control of cancer through external manipulation of the shape of the landscape.[83][84]

In their landmark paper, The Hallmarks of Cancer,[3] Hanahan and Weinberg suggest that cancer can be described by a small number of underlying principles, despite the complexities of the disease. The authors describe how tumor progression proceeds via a process analogous to Darwinian evolution, where each genetic change confers a growth advantage to the cell. These genetic changes can be grouped into six "hallmarks", which drive a population of normal cells to become a cancer. The six hallmarks are:

Genetic instability is defined as an "enabling characteristic" that facilitates the acquisition of other mutations due to defects in DNA repair.

The hallmark "self-sufficiency in growth signals" describes the observation that tumor cells produce many of their own growth signals and thereby no longer rely on proliferation signals from the micro-environment. Normal cells are maintained in a nondividing state by antigrowth signals, which cancer cells learn to evade through genetic changes producing "insensitivity to antigrowth signals". A normal cell initiates programmed cell death (apoptosis) in response to signals such as DNA damage, oncogene overexpression, and survival factor insufficiency, but a cancer cell learns to "evade apoptosis", leading to the accumulation of aberrant cells. Most mammalian cells can replicate a limited number of times due to progressive shortening of telomeres; virtually all malignant cancer cells gain an ability to maintain their telomeres, conferring "limitless replicative potential". As cells cannot survive at distances of more than 100 m from a blood supply, cancer cells must initiate the formation of new blood vessels to support their growth via the process of "sustained angiogenesis". During the development of most cancers, primary tumor cells acquire the ability to undergo "invasion and metastasis" whereby they migrate into the surrounding tissue and travel to distant sites in the body, forming secondary tumors.

The pathways that cells take toward becoming malignant cancers are variable, and the order in which the hallmarks are acquired can vary from tumor to tumor. The early genetic events in tumorigenesis are difficult to measure clinically, but can be simulated according to known biology.[85] Macroscopic tumors are now beginning to be described in terms of their underlying genetic changes, providing additional data to refine the framework described in The Hallmarks of Cancer.

The theory about the monoclonal origin of cancer states that, in general, neoplasms arise from a single cell of origin.[1] While it is possible that certain carcinogens may mutate more than one cell at once, the tumor mass usually represents progeny of a single cell, or very few cells.[1] A series of mutations is required in the process of carcinogenesis for a cell to transition from being normal to pre-malignant and then to a cancer cell.[86] The mutated genes usually belong to classes of caretaker, gatekeeper, landscaper or several other genes. Mutation ultimately leads to acquisition of the ten hallmarks of cancer.

The first malignant cell, that gives rise to the tumor, is often labeled a cancer stem cell.[87]

The cancer stem-cell hypothesis relies on the fact that a lot of tumors are heterogeneous the cells in the tumor vary by phenotype and functions.[87][88][89] Current research shows that in many cancers there is apparent hierarchy among cells.[87][88][89] in general, there is a small population of cells in the tumor about 0.2%1%[88] that exhibits stem cell-like properties. These cells have the ability to give rise to a variety of cells in tumor tissue, self-renew indefinitely, and upon transfer can form new tumors. According to the hypothesis, cancer stem cells are the only cells capable of tumorigenesis initiation of a new tumor.[87] Cancer stem cell hypothesis might explain such phenomena as metastasis and remission.

The monoclonal model of cancer and the cancer stem-cell model are not mutually exclusive.[87] Cancer stem cell arises by clonal evolution as a result of selection for the cell with the highest fitness in the neoplasm. This way, the heterogeneous nature of neoplasm can be explained by two processes clonal evolution, or the hierarchical differentiation of cells, regulated by cancer stem cells.[87] All cancers arise as a result of somatic evolution, but only some of them fit the cancer stem cell hypothesis.[87] The evolutionary processes do not cease when a population of cancer stem cells arises in a tumor. Cancer treatment drugs pose a strong selective force on all types of cells in tumors, including cancer stem cells, which would be forced to evolve resistance to the treatment. Cancer stem cells do not always have to have the highest resistance among the cells in the tumor to survive chemotherapy and re-emerge afterwards. The surviving cells might be in a special microenvironment, which protects them from adverse effects of treatment.[87]

It is currently unclear as to whether cancer stem cells arise from adult stem cell transformation, a maturation arrest of progenitor cells, or as a result of dedifferentiation of mature cells.[88]

Therapeutic resistance has been observed in virtually every form of therapy, from the beginning of cancer therapy.[90] In most cases, therapies appear to select for mutations in the genes or pathways targeted by the drug.

Some of the first evidence for a genetic basis of acquired therapeutic resistance came from studies of methotrexate. Methotrexate inhibits the dihydrofolate reductase (DHFR) gene. However, methotrexate therapy appears to select for cells with extra copies (amplification) of DHFR, which are resistant to methotrexate. This was seen in both cell culture[91] and samples from tumors in patients that had been treated with methotrexate.[92][93][94][95]

A common cytotoxic chemotherapy used in a variety of cancers, 5-fluorouracil (5-FU), targets the TYMS pathway and resistance can evolve through the evolution of extra copies of TYMS, thereby diluting the drug's effect.[96]

In the case of Gleevec (Imatinib), which targets the BCR-ABL fusion gene in chronic myeloid leukemia, resistance often develops through a mutation that changes the shape of the binding site of the drug.[97][98] Sequential application of drugs can lead to the sequential evolution of resistance mutations to each drug in turn.[99]

Gleevec is not as selective as was originally thought. It turns out that it targets other tyrosine kinase genes and can be used to control gastrointestinal stromal tumors (GISTs) that are driven by mutations in c-KIT. However, patients with GIST sometimes relapse with additional mutations in c-KIT that make the cancer cells resistant to Gleevec.[100][101]

Gefitinib(Iressa) and Erlotinib (Tarceva) are epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors used for non-small cell lung cancer patients whose tumors have somatic mutations in EGFR. However, most patients' tumors eventually become resistant to these drugs. Two major mechanisms of acquired resistance have been discovered in patients who have developed clinical resistance to Gefitinib or Erlotinib:[102] point mutations in the EGFR gene targeted by the drugs,[103] and amplification of MET, another receptor tyrosine kinase, which can bypass EGFR to activate downstream signaling in the cell. In an initial study, 22% of tumors with acquired resistance to Gefitinib or Erlotinib had MET amplification.[104] To address these issues, clinical trials are currently assessing irreversible EGFR inhibitors (which inhibit growth even in cell lines with mutations in EGFR), the combination of EGFR and MET kinase inhibitors, and Hsp90 inhibitors (EGFR and MET both require Hsp90 proteins to fold properly). In addition, taking repeated tumor biopsies from patients as they develop resistance to these drugs would help to understand the tumor dynamics.

Selective estrogen receptor modulators (SERMs) are a commonly used adjuvant therapy in estrogen-receptor positive (ER+) breast cancer and a preventive treatment for women at high risk of the disease. There are several possible mechanisms of SERM resistance, though the relative clinical importance of each is debated. These include:[105][106]

Most prostate cancers derive from cells that are stimulated to proliferate by androgens. Most prostate cancer therapies are therefore based on removing or blocking androgens. Mutations in the androgen receptor (AR) have been observed in anti-androgen resistant prostate cancer that makes the AR hypersensitive to the low levels of androgens that remain after therapy.[111] Likewise, extra copies of the AR gene (amplification) have been observed in anti-androgen resistant prostate cancer.[112] These additional copies of the gene are thought to make the cell hypersensitive to low levels of androgens and so allow them to proliferate under anti-androgen therapy.

Resistance to radiotherapy is also commonly observed. However, to date, comparisons of malignant tissue before and after radiotherapy have not been done to identify genetic and epigenetic changes selected by exposure to radiation. In gliomas, a form of brain cancer, radiation therapy appears to select for stem cells,[113][114] though it is unclear if the tumor returns to the pre-therapy proportion of cancer stem cells after therapy or if radiotherapy selects for an alteration that keeps the glioma cells in the stem cell state.

Cancer drugs and therapies commonly used today are evolutionary inert and represent a strong selection force, which leads to drug resistance.[115] A possible way to avoid that is to use a treatment agent that would co-evolve alongside cancer cells.

Anoxic bacteria could be used as competitors or predators in hypoxic environments within tumors.[115] Scientists have been interested in the idea of using anoxic bacteria for over 150 years, but until recently there has been little progress in that field. According to Jain and Forbes, several requirements have to be met by the cells to qualify as efficient anticancer bacterium:[116]

In the process of the treatment, cancer cells are most likely to evolve some form of resistance to the bacterial treatment. However, being a living organism, bacteria would coevolve with tumor cells, potentially eliminating the possibility of resistance.[116]

Since bacteria prefer an anoxic environment, they are not efficient at eliminating cells on the periphery of the tumor, where oxygen supply is efficient. A combination of bacterial treatment with chemical drugs will increase chances of destroying the tumor.[116]

Oncolytic viruses are engineered to infect cancerous cells. Limitations of that method include immune response to the virus and the possibility of the virus evolving into a pathogen.[115]

By manipulating the tumor environment, it is possible to create favorable conditions for the cells with least resistance to chemotherapy drugs to become more fit and outcompete the rest of the population. The chemotherapy, administered directly after, should wipe out the predominant tumor cells.[115]

Mapping between common terms from cancer biology and evolutionary biology:

Read this article:
Somatic evolution in cancer - Wikipedia

Stem Cells Therapy for Autism: Does it Work?

Most of us are familiar with the scientific fact that any living, breathing animal, insect etc. is made up of cells. These cells form tissues and organs that support the existence of the host. Many of us have also heard of stem cells therapy for autism but are unsure about its validity.

Scientists have studied the underlying mechanism of cells, as well as their functioning, and have discovered ways of using the cells to improve the lives of humans and treat diseases. To do so, scientists have discovered stem cells; think of it as the building blocks of a fully differentiated cell.

Stem cells are human cells that can be developed and differentiated into other cell types. These cells can be derived from any part of the body, for example, stem cells from the brain, muscle, bone marrow, etc. Stem cells are versatile in that they can be used to fix damaged tissues. The two essential characteristics of stem cells include: Firstly, the ability to self-renew to create successors identical to the original cell. Secondly, stem cells, unlike cancer cells, are controlled and highly regulated, therefore, stem cells need to be able to give rise to specialized cell types that become part of the healthy body.

The purpose of stem cell therapy is to regenerate and repair damaged tissues and cells in the body. There are two main classes of stem cells. Pluripotent stem cells have the potential to become any cell in the adult body and multipotent stem cells are much more restricted to a specific population or lineage of cells. Other stem cell types include totipotent and unipotent.

Lets look at pluripotent and multipotent stem cells in detail.

Pluripotent stem cells are generated from somatic cells. These mainly come from embryos and, as such, theyre often referred to as embryonic stem cells.

Lets discuss three types of embryonic stem cells that are used to generate pluripotent cells. These include true embryonic stem cells (ES), nuclear transfer of somatic cells (ntES), and parthenogenetic embryonic stem cells (these are stem cells from unfertilized eggs).

The true embryonic stem cells are made from unused embryos, such as those that undergo IVF (in vitro fertilization). The process of IVF is such that the eggs and sperm are fertilized in a lab dish. What then happens is that, through this process, more embryos are generated, usually more than the couple actually need. Those that arent used can be donated to science.

Pluripotent cells made from these unused embryos are not genetically matched to the original hosts. These are mainly used in science for studies to learn how stem cells regenerate.

Every cell contains an organelle called a nucleus. The nucleus contains all the cells genetic information essential to its function. The word somatic refers to any cell in the body.

The process of somatic cell nuclear transfer (SCNT) extracts the nucleus from a somatic cell and transfers it into another cell that has had its own nucleus removed; i.e. the nucleus from the previous cell is being transferred to an egg cell that does not contain a nucleus (unnucleated).

When the nucleus is transferred to another cell, it activates the process of pluripotent cell generation that reprograms the generation of genes in that cell. The egg then becomes a zygote nucleus or a fertilized egg, the cell then replicates and through it embryonic stem cells are created.

Imagine being able to fertilize an egg without fertilization by sperm. Unusual, but science makes crazy things happen.

Parthenogenesis is the process whereby an unfertilized egg develops an embryo without fertilization. This can be achieved through chemical, physical or combined activation methods.

The parthenogenetic embryonic stem cells have the capacity for infinite proliferation and self-renewal, and maintain the ability to differentiate into one or more specialized types of cell or tissue.

pESCs are especially useful for regenerative medicine, and therefore allow the generation of functional cells that could potentially be used as treatment for many incurable diseases in the future.

Multipotent stem cells are unspecialized cell types that have the ability to self-renew and differentiate into specialized cell types. However, these cells are specific to the type of tissue or organ. For example, a multipotent adult stem cell from the bone marrow can become specialized to produce all blood cell types; and cells in the stem cells from neural networks in the brain can specialize to glial and neuronal cells.

When we talk about all blood cell types, we have to get a little scientific, but for the curious mind, all blood cell types refers to platelets, B and T lymphocytes, natural killer cells, dendritic cells.the list goes on.

In addition, for the curious mind, various types of stem cells include hematopoietic stem cells (the ones that make blood cell types), mesenchymal stem cells (differentiate into bone, fat, cartilage, muscle, and skin), and neural stem cells (from neural networks).

Now that weve covered the types of stem cells, the question remains, can stem cell therapy cure autism? Lets have a look.

To answer this question, I refer to the review by Price (2020) as it is the latest up-date data on this subject. It is important to note however, that at the time of reading this article there may be other research data published on this topic.

Several research studies cite immune dysfunction as the cause and effect of autism spectrum disorder (ASD). By virtue of this analogy, it has informed the basis of the stem cell therapy approach for treating autism. This is founded on the properties that regulate the immune system (immuno-regulatory properties).

From the review, it was also found that when exposed to inflammatory stimuli, this may lead to the development of postnatal diagnosis of ASD. Inflammation to the cell describes the process that occurs when the cell is exposed to harmful stimuli such as bacteria, trauma, toxins, heat, and pathogens. The affected cells then release chemicals that cause blood vessels to leak fluid into the tissues, causing swelling.

Therefore, an inflammatory stimuli is that which influences the occurrence of an inflammatory response.

Other bodies of research found an altered level of proteins called cytokines which are essential for interaction and communication between cells in ASD. These may also be the cause of the development of autism spectrum disorder. Some genetic studies propose an association between a genetic loci (a specific point on the genome of the autistic individual) and ASD whose function is related to immune function. While others suggest a possible anomaly in the neuronal signaling pathway that directs communication and information transfer between neurons

All these are proposed reasons that hypothesize the use of stem cell therapy to treat autism biologically. However, all these propositions do not lead to one voice, there are too many hypotheses that make it difficult to narrow down the target area that would potentially treat autism or autism symptoms. Keeping in mind that autism traits are diverse, therefore, narrowing this information down to one plausible pathology is an even greater challenge.

So, is stem cell therapy effective? The answer to this is unknown.

Is ASD caused by genetic, immune dysfunction, or inflammatory stimuli? The answer to this is not clear and theres a vast number of studies that argue different theories.

It is even more disturbing to consider these hypotheses because, for example, each person can experience bacterial or viral infections, or stress that can impact immune functioning and/or lead to inflammation but were not all on the spectrum. Therefore, we cant say that factors which alter our immune functioning lead to the development of neurodevelopmental conditions.

However, according to Price, the study by Riordan et al. (2019) proposes the influence of cytokines for the treatment of autism.The data proposed could be a point in a positive direction to answering whether stem cell therapy could potentially treat autism symptoms.

Unfortunately, there is no data to positively state the effectiveness of stem cell therapy for treating autism. As more research is developed in this field, theres hope that more understanding of autism will arise, and perhaps an alternative form of treatment of autism symptoms can be developed. It is also worth noting the possibility of genetic markers that could help diagnose autism during pregnancy or during the prenatal development stage.

The studies highlighted in this article are simply preliminary assessments. Further research needs to be conducted in order to understand the potential of cell therapies for treating autism.

The findings of these studies vary in hypothesis and this makes generalization hard. Science has developed greatly over years, therefore, for those that believe in the potential of science and all that it could offer, theres a reason to hope that stem cell therapy could potentially be used as treatment for autism in the near future.

Biehl, J. K., & Russell, B. (2009). Introduction to stem cell therapy. The Journal of cardiovascular nursing, 24(2), 98105. https://doi.org/10.1097/JCN.0b013e318197a6a5

Price, J.(2020). Cell therapy approaches to autism: a review of clinical trial data. Molecular Autism, 11, 37 . https://doi.org/10.1186/s13229-020-00348-z

http://stemcell.childrenshospital.org/about-stem-cells/adult-somatic-stem-cells-101/where-do-we-get-adult-stem-cells/

Thermo Fisher Scientific. An Overview of Pluripotent and Multipotent Stem Cell Targets. https://www.thermofisher.com/za/en/home/life-science/antibodies/antibodies-learning-center/antibodies-resource-library/antibody-methods/pluripotent-multipotent-stem-cell-targets.html

Yu, Z., Han, B. (2016). Advantages and limitations of the parthenogenetic embryonic stem cells in cell therapy. Journal of Reproduction and Contraception, 27 (2), Issue 2, 118-124. https://doi.org/10.7669/j.issn.1001-7844.2016.02.0118

Support Autism Parenting Magazine

We hope you enjoyed this article. In order to support us to create more helpful information like this, please consider purchasing a subscription to Autism Parenting Magazine.

Where shall we send the PDF?

Enter you email address below to download your FREE guide & receive top autism parenting tips direct to your inbox

Read more here:
Stem Cells Therapy for Autism: Does it Work?

Long-Lasting Effects: Spaceflight Linked to an Increased Risk of Cancer and Heart Disease – SciTechDaily

The researchers believe it is important to continuously screen the blood of astronauts throughout their careers and during their retirement in order to monitor their health.

A groundbreaking study from the Icahn School of Medicine at Mount Sinai found that astronauts are more likely to develop mutations, potentially connected to spaceflight, that raise astronauts lifelong risk of acquiring cancer and heart disease.

Researchers took blood samples from astronauts who served on space shuttle missions between 1998 and 2001 for the National Aeronautics and Space Administration (NASA). All 14 astronauts were found to have DNA alterations, or somatic mutations, in the blood-forming system (hematopoietic stem cells). Their research, which was recently published in the journalCommunications Biology, raises the possibility that these mutations may be brought on by spaceflight and highlights the importance of routine blood testing for astronauts throughout their careers and during retirement to keep an eye on their health.

Somatic mutations are mutations that occur after conception and in cells other than sperm or egg cells, meaning they cannot be passed on to children. The mutations uncovered in this study were characterized by an excess of blood cells produced from a single clone, a process known as clonal hematopoiesis (CH). Such mutations are commonly brought on by environmental causes, such as exposure to UV radiation or certain chemicals, and may develop as a consequence of chemotherapy or radiation treatment for cancer. There are few symptoms or signs of CH; most individuals are identified via genetic testing of their blood for other disorders. Although CH is not always a sign of disease, it is linked to an increased risk of blood cancer and cardiovascular disease.

An infographic describing the research process. Credit: Communications Biology / Mount Sinai Health System

Astronauts work in an extreme environment where many factors can result in somatic mutations, most importantly space radiation, which means there is a risk that these mutations could develop into clonal hematopoiesis. Given the growing interest in both commercial spaceflights and deep space exploration, and the potential health risks of exposure to various harmful factors that are associated with repeated or long-duration exploration space missions, such as a trip to Mars, we decided to explore, retrospectively, somatic mutation in the cohort of 14 astronauts, said the studys lead author David Goukassian, MD, Professor of Medicine (Cardiology) with the Cardiovascular Research Institute at Icahn Mount Sinai.

The study subjects were astronauts who flew relatively short (median 12 days) space shuttle missions between 1998 and 2001. Their median age was approximately 42 years old; roughly 85 percent were male, and six of the 14 were on their first mission. The researchers collected whole blood samples from the astronauts 10 days before their flight and on the day of landing, and white blood cells only three days after landing. The samples were stored at -80C for approximately 20 years.

Using DNA sequencing followed by extensive bioinformatics analyses, researchers identified 34 mutations in 17 CH-driver genes. The most frequent mutations occurred in TP53, a gene that produces a tumor-suppressing protein, and DNMT3A, one of the most frequently mutated genes in acute myeloid leukemia. However, the frequency of the somatic mutations in the genes that the researchers assessed was less than two percent, the technical threshold for somatic mutations in hematopoietic stem cells to be considered clonal hematopoiesis of indeterminate potential (CHIP). CHIP is more common in older individuals and is associated with an increased risk of developing cardiovascular disease and both hematologic and solid cancer.

Although the clonal hematopoiesis we observed was of relatively small size, the fact that we observed these mutations was surprising given the relatively young age and health of these astronauts. The presence of these mutations does not necessarily mean that the astronauts will develop cardiovascular disease or cancer, but there is the risk that, over time, this could happen through ongoing and prolonged exposure to the extreme environment of deep space, Dr. Goukassian said. Through this study, we have shown that we can determine the individual susceptibility of astronauts to develop disease related to their work without any implications that can affect their ability to do their work. Indeed, our studies demonstrate the importance of early and ongoing screening to assess that susceptibility. Our recommendation is that NASA, and its medical team, screen astronauts for somatic mutations and possible clonal expansion, or regression, every three to five years, and, not less importantly, well into their retirement years when somatic mutations may expand clonally and become CHIP.

The teams research follows previous studies that used the same samples to identify predictive biomarkers in exosomessmall lipid-layered microscopic vesicles of nucleic acids, proteins, lipids, and metabolites that form within the cells of the human body and are subsequently released into the blood circulation, hence carrying the information from their cells of origin that reflects their intercellular condition. This feature of exosomes may qualify them as great biomarkers of health and/or disease, as well as transfer information from one cell to another at a great distance in the body. When they treated human heart cells with exosomes derived from astronauts, the researchers found that the exosomes affected the biology of the vitamin D receptor, which plays a key role in bone, heart, and skeletal muscle health. They also assessed the impact of space flight on mitochondrial DNAthe genome of small organelles that supply energy to cells. In that study, the team found that the amount of cell-free mitochondrial DNA circulating in the blood of astronauts was two to 350 times higher than normal, which may lead to oxidative damage and inflammation elsewhere in the body.

Through these studies, we have demonstrated the potential to assess the health risk of space flight among astronauts. What is important now is to conduct longitudinal retrospective and well-controlled prospective studies involving a large number of astronauts to see how that risk evolves based on continued exposure and then compare that data against their clinical symptoms, imaging, and lab results. That will enable us to make informed predictions as to which individuals are more likely to develop disease based on the phenomena we are seeing and open the door to individualized precision medicine approaches to early intervention and prevention, said Dr. Goukassian.

Reference: Retrospective analysis of somatic mutations and clonal hematopoiesis in astronauts by Agnieszka Brojakowska, Anupreet Kour, Mark Charles Thel, Eunbee Park, Malik Bisserier, Venkata Naga Srikanth Garikipati, Lahouaria Hadri, Paul J. Mills, Kenneth Walsh and David A. Goukassian, 17 August 2022, Communications Biology. DOI: 10.1038/s42003-022-03777-z

See the rest here:
Long-Lasting Effects: Spaceflight Linked to an Increased Risk of Cancer and Heart Disease - SciTechDaily

Global Induced Pluripotent Stem Cells Market (2022 to 2027) – Growth, Trends, Covid-19 Impact and Forecasts – ResearchAndMarkets.com – Business Wire

DUBLIN--(BUSINESS WIRE)--The "Induced Pluripotent Stem Cells Market - Growth, Trends, Covid-19 Impact, and Forecasts (2022 - 2027)" report has been added to ResearchAndMarkets.com's offering.

The Induced Pluripotent Stem Cells Market is projected to register a CAGR of 8.4% during the forecast period (2022 to 2027).

Companies Mentioned

Key Market Trends

The Drug Development Segment is Expected to Hold a Major Market Share in the Induced Pluripotent Stem Cells Market.

By application, the drug development segment holds the major segment in the induced pluripotent stem cell market. Various research studies focusing on drug development studies with induced pluripotent stem cells have been on the rise in recent years.

For instance, an article titled "Drug Development and the Use of Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Disease Modeling and Drug Toxicity Screening" published in the International Journal of Molecular Science in October 2020 discussed the broad use of iPSC derived cardiomyocytes for drug development in terms of adverse drug reactions, mechanisms of cardiotoxicity, and the need for efficient drug screening protocols.

Another article published in the Journal of Cells in December 2021 titled "Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective" focused on methods to reprogram somatic cells into human induced pluripotent stem cells and the solutions to overcome the immaturity of the human induced pluripotent stem cells derived cardiomyocytes to mimic the structure and physiological properties of adult human cardiomyocytes to accurately model disease and test drug safety. Thus, this increase in the research of induced pluripotent stem cells for drug development and drug modeling is likely to propel the segment's growth over the study period.

Furthermore, as per an article titled "Advancements in Disease Modeling and Drug Discovery Using iPSC-Derived Hepatocyte-like Cells" published in the Multi-Disciplinary Publishing Institute journal of Cells in March 2022, preserved differentiation and physiological function, amenability to genetic manipulation via tools such as CRISPR/Cas9, and availability for high-throughput screening, make induced pluripotent stem cell systems increasingly attractive for both mechanistic studies of disease and the identification of novel therapeutics.

North America is Expected to Hold a Significant Share in the Market and Expected to do Same in the Forecast Period

The rise in the adoption of highly advanced technologies and systems in drug development, toxicity testing, and disease modeling coupled with the growing acceptance of stem cell therapies in the region are some of the major factors driving the market growth in North America.

The United States Food and Drug Administration in March 2022 discussed the development of strategies to improve cell therapy product characterization. The agency focused on the development of improved methods for testing stem cell products to ensure the safety and efficacy of such treatments when used as therapies.

Likewise, in March 2020, the Food and Drug Administration announced that ImStem drug IMS001, which uses AgeX's pluripotent stem cell technology, would be available for the treatment of multiple sclerosis. Similarly, REPROCELL introduced a customized iPSC generation service in December 2020, as well as a new B2C website to promote the "Personal iPS" service. This service prepares and stores an individual's iPSCs for future injury or disease regeneration treatment.

Thus, the increasing necessity for induced pluripotent stem cells coupled with increasing investment in the health care department is known to propel the growth of the market in this region.

Key Topics Covered:

1 INTRODUCTION

2 RESEARCH METHODOLOGY

3 EXECUTIVE SUMMARY

4 MARKET DYNAMICS

4.1 Market Overview

4.2 Market Drivers

4.2.1 Increase in Research and Development Activities in Stem Cells Therapies

4.2.2 Surge in Adoption of Personalized Medicine

4.3 Market Restraints

4.3.1 Lack of Awareness Regarding Stem Cell Therapies

4.3.2 High Cost of Treatment

4.4 Porter's Five Force Analysis

5 MARKET SEGMENTATION

5.1 By Derived Cell Type

5.2 Application

5.3 End User

5.4 Geography

6 COMPETITIVE LANDSCAPE

6.1 Company Profiles

7 MARKET OPPORTUNITIES AND FUTURE TRENDS

For more information about this report visit https://www.researchandmarkets.com/r/ylzwhr

Read the original:
Global Induced Pluripotent Stem Cells Market (2022 to 2027) - Growth, Trends, Covid-19 Impact and Forecasts - ResearchAndMarkets.com - Business Wire

Utilization of Modified Induced Pluripotent Stem Cells as the Advance | OPTH – Dove Medical Press

Introduction

Glaucoma is one of the optic neuropathy disorders characterized by the progressive degeneration of retinal ganglion cells (RGC), which eventually lead to cupping of the optic disc and decreased visual field.1 Glaucoma is also closely related to an increase in intraocular pressure caused by the damage of trabecular meshwork (TBM), which results in optic nerve damage, characterized by the loss of retinal ganglion cells.2,3 Globally, in 2020, more than 76 million people are suffering from glaucoma, and it is expected to increase to 111.8 million people by 2040.4,5 Glaucoma is also a severe and complex medical problem because it often causes blindness. According to the World Health Organization (WHO), the most common causes of blindness are cataracts (51%), followed by glaucoma (8%), and age-related macular degeneration (5%).6 This data shows that glaucoma is the worlds second most common cause of blindness after cataracts. Symptoms that are often asymptomatic at an early stage and the low public awareness have contributed to the disorders seriousness.

Handling and treating glaucoma cases is difficult, especially because no therapy can cure glaucoma. Current treatment, both medical and surgical, is focused solely on lowering intraocular pressure. Treatment of glaucoma cases should also be carried out for life to maintain normal intraocular pressure and prevent the progression of intraocular damage due to glaucoma.7 Based on these problems, innovation is needed to handle glaucoma effectively. Besides, solutions are also required to repair the damage to retinal ganglion cells in glaucoma. One of the therapies that researchers are trying to take advantage of is stem cell therapy, a technology where cells can develop into many specific cells desired.8 In cases of glaucoma or optic neuropathies, damaged RGCs can be replaced with new ones grown from stem cells.9 Another option for RGC regeneration is to use retinal stem cells to regenerate RGCs. Indeed, stem cell therapy relies on exogenous stem cell sources due to their limited availability. Currently, many stem cell therapies for eye diseases that are created and studied are limited to treating the damage of photoreceptors and retinal pigment epithelium. iPSC-derived RGCs can serve as an excellent model for formulating approaches to promote de novo-generated RGCs to connect with their targets. Therefore, researchers have been looking into the potential use of modified stem cell therapy to treat the intraocular injury in glaucoma cases.10

This review aims to synthesize and prove the efficacy and further modification of this method so that it can be eligible for treatment and can also give data collection for the scientific community. This systematic review is expected to provide detailed information regarding the possible applications of modified stem cell therapy in treating intraocular damage in glaucoma patients.

In the present literature review, literature regarding the potential utilization of stem cells as an advanced therapy for intraocular glaucomatous damage was searched. The stages of this literature review include five steps: i) identifying the research question, ii) identifying relevant studies, iii) study selection, iv) charting the data, and v) summarizing and reporting the results.

This literature review was conducted to answer the following research questions:

The literature search was carried out from January to February 2021. Keywords and synonyms used to conduct literature searches related to the research question are attached in Table 1. Boolean operators (OR, AND, NOT) combine keywords when searching for literature. The search was conducted on seven online databases, namely PubMed, ScienceDirect, ProQuest, EBSCOhost, SAGE, Clinicalkey, and Scopus.

Table 1 Keywords That Were Used in the Database Search

The inclusion criteria for the literature search consisted of journals published in English and journals published in the last ten years. The exclusion criteria for selected studies consisted of journals that were not fully accessible due to the limited facilities owned as supporting access. We thoroughly screened the titles and abstracts of the studies obtained to suit the purpose of this literature review. Abstracts that were not relevant to the research objectives were excluded. Then a full article screening was carried out from the selected abstracts to identify whether the full article was suitable for the research objectives and whether the full article could be used to answer research questions.

Information obtained from all selected study articles is then displayed in the charting table The information displayed includes the author, year of publication, study objectives, location, study design, inclusion and exclusion criteria, results, and conclusions.

The researcher did not assess the quality of the selected articles because this study was only a literature review. The data from selected studies are reported to produce recommendations for further research regarding the use of stem cell therapy in glaucoma cases.

Based on the literature search that has been conducted, a total of 2262 studies and abstracts were included in the journal screening process at an early stage. From this screening process, 362 duplicate articles were excluded from the selection. The remaining 1900 articles then entered the abstract eligibility screening stage. Only 53 articles were selected, while 1879 other articles were excluded. Of the 53 articles, 18 articles appeared relevant to the study and met the inclusion criteria for review throughout the study. Meanwhile, 35 other studies were excluded because the focus in these studies did not match the objectives of this literature review. After assessing the full articles, six studies met the inclusion criteria in this literature review (Figure 1).

Figure 1 Flow diagram of the literature review process.

In Table 2, a summary of the characteristics of the selected studies is presented. The data used from selected studies include research objectivity, study design, results, outputs, and conclusions from the study. Of all the selected studies, there were six studies that had experimental methods. Almost all studies have the aim of evaluating and proving the potential of using stem cells to replace damaged tissue and restore and restore the function of damaged eye tissue, particularly due to degenerative processes such as disease of the retina or glaucoma.

Table 2 Results Summary of the Characteristics of the Selected Studies

Glaucoma is characterized by the degeneration of retinal ganglion cells. Based on the pathophysiology, glaucoma can be divided into two categories, namely open-angle glaucoma and closed-angle glaucoma. In patients with open-angle glaucoma, there is increased resistance to the aqueous humors outflow through the trabecular meshwork. This increased resistance is often caused by apoptosis and senescence of trabecular meshwork cells with increasing age.15 Degradation and abnormalities of the cytoskeleton arrangement of trabecular meshwork cells resulting in thickening of the drainage pathways and abnormal extracellular matrix deposition also worsen trabecular meshwork function in open-angle glaucoma.16 In closed-angle glaucoma, the aqueous humor cannot reach the trabecular meshwork due to obstruction.17 Examples of obstructions that often cause closed-angle glaucoma are anterior synechiae, the attachment of the iris to the trabecular meshwork, and posterior synechiae, where the iris is attached to the lens. This adhesion causes the aqueous humor to fail to reach the drainage system and the trabecular meshwork.18

Glaucoma is closely related to increased intraocular pressure, which is determined by the balance between the production of aqueous humor by the ciliary body and the drainage of the aqueous humor through the trabecular meshwork. The disturbance of the balance between production and drainage increases the humor Aquos, which at a later stage can increase the intraocular pressure.19 Studies have shown a link between increased intraocular pressure and retinal ganglion cell death. This study has also proven that the longer the intraocular pressure increases, the higher the degree of retinal ganglion cell damage.20 However, data show as many as 3040% of patients with glaucoma have normal intraocular pressure. One of the causes of glaucoma at normal intraocular pressure is a decrease in neurotrophic factors needed in the maintenance of neurons in the optic nerve. Neurotrophic factors are required to maintain retinal ganglion cells, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and cell line-derived neurotrophic factor.21 Furthermore, microcirculation disorders, changes in immune system conditions, and increased levels of oxidative stress can also cause glaucoma at normal intraocular pressure.21

Stem cells are cells with the ability to differentiate and form all tissues in the human body. They are one of the potential therapies used in cases that require tissue repair and regeneration, one of which is glaucoma. For a cell to be called a stem cell, it must have two essential characteristics. The first one is the stem cell must produce offspring with the exact features the cell originates from, and the second one, the stem cell must be able to differentiate into the specific cell desired.22 There are two types of stem cells found in multicellular organisms, including humans. The first stem cells are embryonic stem cells or multipotent cells found in blastocysts, while the second stem cells are adult stem cells or pluripotent cells that can be found in a wide variety of adult tissues.23

Research has also succeeded in inducing adult cells to return to the pluripotent stage using molecular manipulation. The cells produced by this molecular manipulation are then called induced pluripotent stem cells (iPS).24 Most iPS manufacturing uses viruses such as retroviruses and lentiviruses to carry genes encoding transcription factors to adult cells to be modified. This gene will then undergo transcription and translation into a protein capable of inducing the adult cell nucleus to return to an embryonic state.25

An important concept that needs attention in stem cell therapy is how to induce stem cells to become the desired differentiated cells.26 It is necessary so that the cells can be used to treat various diseases, including glaucoma. We can further achieve differentiation of stem cells into specific desired cells by adding various growth factors and signaling pathways to resemble the conditions of their original development.27

The research conducted successfully isolates cultures and confirms that the trabecular meshwork stem cells around the Schwalbe line are multipotent with the ability to differentiate into a wide variety of cells, including trabecular meshwork cells adipocytes osteocytes, and chondrocytes.28 Other studies have also been able to induce stem cells on the Schwalbe line trabecular meshwork to proliferate and differentiate into photoreceptors under certain conditions.29 Apart from trabecular meshwork stem cells, other stem cells that can differentiate into functional meshwork trabecular cells are adipose-derived stem cells (ADSC), mesenchymal stem cells (MSC), and iPS. iPS cells can also differentiate into trabecular meshwork cells after culturing the extracellular matrix with cell-derived trabecular meshwork. The success of a wide variety of stem cells to differentiate into functional meshwork trabecular cells provides a more effective alternative to cutting-edge therapy in treating glaucoma, especially open-angle glaucoma.3

One of the stem cell therapies successfully applied and able to regenerate damaged retinal ganglion cells is iPS cell therapy. This therapy uses induced adult fibroblasts to return to pluripotent cells using four transcription factors, namely Oct3/4, Sox2, Klf4, and c-Myc. The results of the iPS are pluripotent cell colonies that are morphologically similar to ESCs, which are able to differentiate into the three germ cell layers.30

Because iPS can be programmed from the patients somatic cells, this therapy can maintain the unique genome of each individual. Currently, various modifications to the iPS therapy have been made to increase its acceptability and effectiveness of iPS therapy. One of them is the use of plasmid vectors and miRNA instead of retroviruses to avoid mutagenesis of the adult cells used.31,32

One of the significant challenges in stem cell therapy is to achieve the differentiation of stem cells into the desired cells, in this case, the differentiation of stem cells to retinal ganglion cells. Usually, in vivo, the differentiation of stem cells into retinal ganglion cells is regulated by several transcription factors such as Ath5, Brn3, and Notch. The transcription factors Ath5 and Brn3 play a vital role in the differentiation of retinal ganglion cells, and their levels are increased in the process of eye development.33 Meanwhile, Notch is a negative regulator of retinal ganglion cell differentiation, and its levels are decreased in normal eye development. Therefore, the addition of the transcription factors Ath5 and Brn3 and the Notch antagonist is a strategy to differentiate retinal ganglion cells from stem cells.34 Apart from transcription factors, various neurotrophic pathways and factors have been identified in the differentiation of stem cells into retinal ganglion cells. These pathways consist of fibroblast growth factor (FGF), insulin-like growth factor (IGF), bone morphogenetic protein (BMP), nodal, and Wnt signaling pathways. All of these pathways regulate retinal development, whereas FGF and IGF provide positive regulation. Meanwhile, BMP, nodal, and Wnt signaling pathways provide negative regulation.35

Another major challenge in the clinical application of stem cell therapy in glaucoma sufferers is that not only do the stem cells successfully differentiate into retinal ganglion cells, but they must also be able to reach the central nervous system.36 Modifications must be made so that new retinal ganglion cells can reach the visual cortex of the cerebrum. Recent research has found that a combination of genetic modification and stimulation of the signaling pathway stimulates regeneration of the optic nerve until it reaches the central nervous system. The addition of ephrin molecules, proteoglycans, cell-adhesion molecules, and semaphorin is able to guide the axons of the developing retinal ganglion cells to reach the optic chiasm.13 Meanwhile, the addition of cadherin, ephrin, and the Wnt signaling pathway can guide and stimulate synapse formation in the superior colliculus and the visual cortex.12,37

In addition, because of the adverse intraocular environment in glaucoma, stem cell therapy needs to be combined with neuroprotective compounds. It is also associated with a decrease in neurotrophic factors required to maintain neurons and causes progression of retinal ganglion cell damage in glaucoma sufferers. Therefore, the addition of BDNF and other neurotrophic factors such as glial cell-derived neurotrophic factor (GDNF) and ciliary neurotrophic factor (CNTF) should be considered for combined stem cell therapy.38

The stem cells are used in cases of glaucoma, which require repair and regeneration of trabecular meshwork cells and retinal ganglion cells. iPS has been shown the ability to differentiate to replace damaged trabecular meshwork cells and retinal ganglion cells in glaucoma. Some modifications are required so that stem cells that have differentiated into trabecular meshwork cells and retinal ganglion cells can reach the central nervous system. These modifications include the addition of ephrin molecules, proteoglycans, cell-adhesion molecules, semaphorin, cadherin, and the Wnt signaling pathway. The combination of stem cells with neuroprotective factors such as BDNF, GDNF, and CNTF also needs to be considered to maintain neuronal maintenance and inhibit the progression of cell damage.

The development of new stem cell technologies not only paves the way for us to gain a better understanding of the biology associated with glaucoma and create models for the development of new drugs, but it also opens the door to the prospect of cell-based therapies that can help patients regain their vision. More specifically in relation to the field of glaucoma, there have been recent developments in the process of developing protocols for the differentiation of stem cells into trabecular meshwork and retinal ganglion cells. Further research on the effectiveness of using modified stem cells as a therapy for glaucoma and in vivo research can be carried out immediately so that clinical trials can be carried out, which in turn can be used by the community to control symptoms and reduce blindness due to glaucoma.

The authors report no conflicts of interest in this work.

1. Choudhari NS, Neog A, Fudnawala V, George R. Cupped disc with normal intraocular pressure: the long road to avoid misdiagnosis. Indian J Ophthalmol. 2011;59(6):491. doi:10.4103/0301-4738.86320

2. Braunger BM, Ademoglu B, Koschade SE, et al. Identification of adult stem cells in Schwalbes line region of the primate eye. Invest Ophthalmol Vis Sci. 2014;55(11):7499. doi:10.1167/iovs.14-14872

3. Abu-Hassan DW, Li X, Ryan EI, Acott TS, Kelley MJ. Induced pluripotent stem cells restore function in a human cell loss model of open-angle glaucoma. Stem Cells. 2015;33(3):751761. doi:10.1002/stem.1885

4. Cook C, Foster P. Epidemiology of glaucoma: whats new? Can J Ophthalmol. 2012;47(3):223226. doi:10.1016/j.jcjo.2012.02.003

5. Hapsari DM. The Relationship Between Family Support and Adherence in Care in Glaucoma Clients in the Working Area of the Balung Health Center. Jember Regency. Uni; 2017.

6. World Health Organization. Global data on visual impairments 2010; 2012. https://www.iapb.org/wp-content/uploads/GLOBALDATAFINALforweb.pdf. Accessed August 24, 2022.

7. Harasymowycz P, Birt C, Gooi P, et al. Medical management of glaucoma in the 21st century from a canadian perspective. J Ophthalmol. 2016;2016:122. doi:10.1155/2016/6509809

8. Romito A, Cobellis G. Pluripotent stem cells: current understanding and future directions. Stem Cells Int. 2016;2016:120. doi:10.1155/2016/9451492

9. Cen L-P, Ng TK. Stem cell therapy for retinal ganglion cell degeneration. Neural Regen Res. 2018;13(8):1352. doi:10.4103/1673-5374.235237

10. ner A. Stem cell treatment in retinal diseases: recent developments. Turk J Ophthalmol. 2018;48(1). doi:10.4274/tjo.89972

11. Manuguerra-Gagn R, Boulos PR, Ammar A, et al. Transplantation of mesenchymal stem cells promotes tissue regeneration in a glaucoma model through laser-induced paracrine factor secretion and progenitor cell recruitment. Stem Cells. 2013;31(6):11361148. doi:10.1002/stem.1364

12. Osterhout JA, Josten N, Yamada J, et al. Cadherin-6 mediates axon-target matching in a non-image-forming visual circuit. Neuron. 2011;71(4):632639. doi:10.1016/j.neuron.2011.07.006

13. De lima S, Koriyama Y, Kurimoto T, et al. Full-length axon regeneration in the adult mouse optic nerve and partial recovery of simple visual behaviors. Proc Natl Acad Sci U S A. 2012;109(23):91499154. doi:10.1073/pnas.1119449109

14. Deng F, Chen M, Liu Y, et al. Stage-specific differentiation of iPSCs toward retinal ganglion cell lineage. Mol Vis. 2016;22:536.

15. Pulliero A, Seydel A, Camoirano A, Sacc SC, Sandri M, Izzotti A. Oxidative damage and autophagy in the human trabecular meshwork as related with ageing. PLoS One. 2014;9(6):e98106. doi:10.1371/journal.pone.0098106

16. Porter KM, Epstein DL, Liton PB. Up-regulated expression of extracellular matrix remodeling genes in phagocytically challenged trabecular meshwork cells. PLoS One. 2012;7(4):e34792. doi:10.1371/journal.pone.0034792

17. Lai J, Choy BNK, Shum JWH. Management of primary angle-closure glaucoma. Asia Pac J Ophthalmol. 2016;5(1):5962. doi:10.1097/APO.0000000000000180

18. Sun X, Dai Y, Chen Y, et al. Primary angle closure glaucoma: what we know and what we dont know. Prog Retin Eye Res. 2017;57:2645. doi:10.1016/j.preteyeres.2016.12.003

19. Gemenetzi M, Yang Y, Lotery AJ. Current concepts on primary open-angle glaucoma genetics: a contribution to disease pathophysiology and future treatment. Eye. 2012;26(3):355369. doi:10.1038/eye.2011.309

20. Khan Kareem A, Tse DY, van der Heijden ME, et al. Prolonged elevation of intraocular pressure results in retinal ganglion cell loss and abnormal retinal function in mice. Exp Eye Res. 2015;130. doi:10.1016/j.exer.2014.11.007

21. Almasieh M, Wilson AM, Morquette B, Cueva Vargas JL, Di Polo A. The molecular basis of retinal ganglion cell death in glaucoma. Prog Retin Eye Res. 2012;31(2):152181. doi:10.1016/j.preteyeres.2011.11.002

22. Cao J, Ng ES, Mcnaughton D, et al. The characterisation of pluripotent and multipotent stem cells using Fourier transform infrared microspectroscopy. Int J Mol Sci. 2013;14(9):1745317476. doi:10.3390/ijms140917453

23. Makhani K, Ali SM, Yousuf S, Siddiqui S. Therapeutic potential of totipotent, pluripotent and multipotent stem cells. MOJ Cell Sci Rep. 2015;2(5):00041. doi:10.15406/mojcsr.2015.02.00041

24. Zaveri L, Dhawan J. Cycling to meet fate: connecting pluripotency to the cell cycle. Front Cell Dev Biol. 2018;6. doi:10.3389/fcell.2018.00057

25. Wattanapanitch M. Recent updates on induced pluripotent stem cells in hematological disorders. Stem Cells Int. 2019;2019:115. doi:10.1155/2019/5171032

26. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2012;85(1):310. doi:10.1159/000345615

27. Biehl JK, Russell B. Introduction to stem cell therapy. J Cardiovasc Nurs. 2009;24(2):98103; quiz 1045. doi:10.1097/JCN.0b013e318197a6a5

28. Tay CY, Sathiyanathan P, Chu SWL, Stanton LW, Wong TT. Identification and characterization of mesenchymal stem cells derived from the trabecular meshwork of the human eye. Stem Cells Dev. 2012;21(9):13811390. doi:10.1089/scd.2011.0655

29. Nadri S, Yazdani S, Arefian E, et al. Mesenchymal stem cells from trabecular meshwork become photoreceptor-like cells on amniotic membrane. Neurosci Lett. 2013;541:4348. doi:10.1016/j.neulet.2012.12.055

30. Omole AE, Fakoya AOJ. Ten years of progress and promise of induced pluripotent stem cells: historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ. 2018;2018. doi:10.7717/peerj.4370

31. Miyoshi N, Ishii H, Nagano H, et al. Reprogramming of mouse and human cells to pluripotency using mature microRNAs. Cell Stem Cell. 2011;8(6):633638. doi:10.1016/j.stem.2011.05.001

32. Ikushima S, Ono R, Fukuda K, Sakayori M, Awano N, Kondo K. Trousseaus syndrome: cancer-associated thrombosis. Jpn J Clin Oncol. 2016;46(3):204208. doi:10.1093/jjco/hyv165

33. Ji SL, Tang SB. Differentiation of retinal ganglion cells from induced pluripotent stem cells: a review. Int J Ophthalmol. 2019;12(1):152160. doi:10.18240/ijo.2019.01.22

34. Li L, Chen LP, Liu QH. Effect of the notch signaling pathway on retinal ganglion cells and its neuroprotection in rats with acute ocular hypertension. Int J Ophthalmol. 2018;11(2). doi:10.18240/ijo.2018.02.05

35. Ritchey ER, Zelinka CP, Tang J, Liu J, Fischer AJ. The combination of IGF1 and FGF2 and the induction of excessive ocular growth and extreme myopia. Exp Eye Res. 2012;99(1):116. doi:10.1016/j.exer.2012.03.019

36. Georgiev D. Photons do collapse in the retina not in the brain cortex: evidence from visual illusions. NeuroQuantology. 2011;9(2). doi:10.14704/nq.2011.9.2.403

37. He CW, Liao CP, Pan CL. Wnt signalling in the development of axon, dendrites and synapses. Open Biol. 2018;8(10):180116. doi:10.1098/rsob.180116

38. Mysona BA, Zhao J, Bollinger KE. Role of BDNF/TrkB pathway in the visual system: therapeutic implications for glaucoma. Expert Rev Ophthalmol. 2017;12(1):6981. doi:10.1080/17469899.2017.1259566

Read the original post:
Utilization of Modified Induced Pluripotent Stem Cells as the Advance | OPTH - Dove Medical Press

Fragment size and dynamics of EGFR-mutated tumor-derived DNA provide prognostic information regarding EGFR-TKI efficacy in patients with EGFR-mutated…

Planchard, D. et al. Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 29, iv192iv237 (2018).

CAS PubMed Google Scholar

Ettinger, D. S. et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, 2020. J. Natl. Compr. Canc. Netw. 17, 14641472 (2019).

PubMed Google Scholar

Aggarwal, C. et al. Strategies for the successful implementation of plasma-based NSCLC genotyping in clinical practice. Nat. Rev. Clin. Oncol. 18, 5662 (2021).

PubMed Google Scholar

Kunimasa, K. et al. Improvement strategies for successful next-generation sequencing analysis of lung cancer. Future Oncol. 16, 15971606 (2020).

CAS PubMed Google Scholar

Leighl, N. B. et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin. Cancer Res. 25, 46914700 (2019).

CAS PubMed Google Scholar

Saarenheimo, J., Andersen, H., Eigeliene, N. & Jekunen, A. Gene-guided treatment decision-making in non-small cell lung cancerA systematic review. Front. Oncol. 11, 754427 (2021).

PubMed PubMed Central Google Scholar

Rolfo, C. et al. Liquid biopsy for advanced NSCLC: A consensus statement from the International Association for the Study of Lung Cancer. J. Thorac. Oncol. 16, 16471662 (2021).

CAS PubMed Google Scholar

Laufer-Geva, S. et al. The clinical impact of comprehensive genomic testing of circulating cell-free DNA in advanced lung cancer. J. Thorac. Oncol. 13, 17051716 (2018).

PubMed Google Scholar

Woodhouse, R. et al. Clinical and analytical validation of FoundationOne Liquid CDx, a novel 324-Gene cfDNA-based comprehensive genomic profiling assay for cancers of solid tumor origin. PLoS ONE 15, e0237802 (2020).

CAS PubMed PubMed Central Google Scholar

Newman, A. M. et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 20, 548554 (2014).

CAS PubMed PubMed Central Google Scholar

Kukita, Y. et al. High-fidelity target sequencing of individual molecules identified using barcode sequences: De novo detection and absolute quantitation of mutations in plasma cell-free DNA from cancer patients. DNA Res. 22, 269277 (2015).

CAS PubMed PubMed Central Google Scholar

Casbon, J. A., Osborne, R. J., Brenner, S. & Lichtenstein, C. P. A method for counting PCR template molecules with application to next-generation sequencing. Nucl. Acids Res. 39, e81 (2011).

CAS PubMed PubMed Central Google Scholar

Kinde, I., Wu, J., Papadopoulos, N., Kinzler, K. W. & Vogelstein, B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl. Acad. Sci. U.S.A. 108, 95309535 (2011).

ADS PubMed PubMed Central Google Scholar

Akahori, D. et al. Comparative assessment of NOIR-SS and ddPCR for ctDNA detection of EGFR L858R mutations in advanced L858R-positive lung adenocarcinomas. Sci. Rep. 11, 14999 (2021).

CAS PubMed PubMed Central Google Scholar

Takada, R. et al. Clinical utility of pancreatic cancer circulating tumor DNA in predicting disease progression, prognosis, and response to chemotherapy. Pancreas 49, e93e95 (2020).

CAS PubMed Google Scholar

Leon, S. A., Shapiro, B., Sklaroff, D. M. & Yaros, M. J. Free DNA in the serum of cancer patients and the effect of therapy. Cancer Res. 37, 646650 (1977).

CAS PubMed Google Scholar

Mouliere, F. et al. High fragmentation characterizes tumour-derived circulating DNA. PLoS ONE 6, e23418 (2011).

ADS CAS PubMed PubMed Central Google Scholar

Yamamoto, Y. et al. Clinical significance of the mutational landscape and fragmentation of circulating tumor DNA in renal cell carcinoma. Cancer Sci. 110, 617628 (2019).

CAS PubMed PubMed Central Google Scholar

Liu, X. et al. Enrichment of short mutant cell-free DNA fragments enhanced detection of pancreatic cancer. EBioMedicine 41, 345356 (2019).

PubMed PubMed Central Google Scholar

Underhill, H. R. et al. Fragment length of circulating tumor DNA. PLoS Genet. 12, e1006162 (2016).

PubMed PubMed Central Google Scholar

Liu, Y. et al. Increased detection of circulating tumor DNA by short fragment enrichment. Transl. Lung Cancer Res. 10, 15011511 (2021).

CAS PubMed PubMed Central Google Scholar

Mouliere, F. et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aat4921 (2018).

Article PubMed PubMed Central Google Scholar

Underhill, H. R. Leveraging the fragment length of circulating tumour DNA to improve molecular profiling of solid tumour malignancies with next-generation sequencing: A pathway to advanced non-invasive diagnostics in precision oncology?. Mol. Diagn. Ther. 25, 389408 (2021).

CAS PubMed PubMed Central Google Scholar

Li, F. et al. Ultra-short circulating tumor DNA (usctDNA) in plasma and saliva of non-small cell lung cancer (NSCLC) patients. Cancers (Basel) 12, 2041 (2020).

CAS Google Scholar

Heitzer, E., Auinger, L. & Speicher, M. R. Cell-free DNA and apoptosis: How dead cells inform about the living. Trends Mol. Med. 26, 519528 (2020).

CAS PubMed Google Scholar

Jahr, S. et al. DNA fragments in the blood plasma of cancer patients: Quantitations and evidence for their origin from apoptotic and necrotic cells. Cancer Res. 61, 16591665 (2001).

CAS PubMed Google Scholar

Jensen, T. J. et al. Whole genome bisulfite sequencing of cell-free DNA and its cellular contributors uncovers placenta hypomethylated domains. Genome Biol. 16, 78 (2015).

PubMed PubMed Central Google Scholar

Udomruk, S., Orrapin, S., Pruksakorn, D. & Chaiyawat, P. Size distribution of cell-free DNA in oncology. Crit. Rev. Oncol. Hematol. 166, 103455 (2021).

PubMed Google Scholar

Park, S. et al. High concordance of actionable genomic alterations identified between circulating tumor DNA-based and tissue-based next-generation sequencing testing in advanced non-small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer 127, 30193028 (2021).

CAS PubMed Google Scholar

Stroun, M., Lyautey, J., Lederrey, C., Olson-Sand, A. & Anker, P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin. Chim. Acta 313, 139142 (2001).

CAS PubMed Google Scholar

Wang, B. G. et al. Increased plasma DNA integrity in cancer patients. Cancer Res. 63, 39663968 (2003).

CAS PubMed Google Scholar

Avanzini, S. et al. A mathematical model of ctDNA shedding predicts tumor detection size. Sci. Adv. https://doi.org/10.1126/sciadv.abc4308 (2020).

Article PubMed PubMed Central Google Scholar

Ying, H. Q. et al. Cancer-elicited inflammation attenuates response and outcome in tyrosine kinase inhibitor naive patients with advanced NSCLC. Pharmacol. Res. 170, 105734 (2021).

CAS PubMed Google Scholar

Chen, Y. M. et al. Impact of clinical parameters and systemic inflammatory status on epidermal growth factor receptor-mutant non-small cell lung cancer patients readministration with epidermal growth factor receptor tyrosine kinase inhibitors. BMC Cancer 16, 868 (2016).

CAS PubMed PubMed Central Google Scholar

Garrido, P. et al. LungBEAM: A prospective multicenter study to monitor stage IV NSCLC patients with EGFR mutations using BEAMing technology. Cancer Med. 10, 58785888 (2021).

CAS PubMed PubMed Central Google Scholar

Sueoka-Aragane, N. et al. The role of comprehensive analysis with circulating tumor DNA in advanced non-small cell lung cancer patients considered for osimertinib treatment. Cancer Med. 10, 38733885 (2021).

CAS PubMed PubMed Central Google Scholar

Sakai, K. et al. Predicting osimertinib-treatment outcomes through EGFR mutant-fraction monitoring in the circulating tumor DNA of EGFR T790M-positive patients with non-small cell lung cancer (WJOG8815L). Mol. Oncol. 15, 126137 (2021).

CAS PubMed Google Scholar

Mok, T. S. et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 376, 629640 (2017).

CAS PubMed Google Scholar

Papadimitrakopoulou, V. A. et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis. Ann Oncol. 31, 15361544 (2020).

CAS PubMed Google Scholar

Oxnard, G. R. et al. Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 4, 15271534 (2018).

PubMed Google Scholar

Wang, F. et al. Genomic temporal heterogeneity of circulating tumour DNA in unresectable metastatic colorectal cancer under first-line treatment. Gut 71, 13401349 (2021).

PubMed Google Scholar

Shaw, A. T. et al. Resensitization to crizotinib by the lorlatinib ALK resistance mutation L1198F. N. Engl. J. Med. 374, 5461 (2016).

CAS PubMed Google Scholar

Blakely, C. M. et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat. Genet. 49, 16931704 (2017).

CAS PubMed PubMed Central Google Scholar

Mouliere, F. et al. Fragmentation patterns and personalized sequencing of cell-free DNA in urine and plasma of glioma patients. EMBO Mol. Med. 13, e12881 (2021).

CAS PubMed PubMed Central Google Scholar

Nagai, Y. et al. Genetic heterogeneity of the epidermal growth factor receptor in non-small cell lung cancer cell lines revealed by a rapid and sensitive detection system, the peptide nucleic acid-locked nucleic acid PCR clamp. Cancer Res. 65, 727682 (2005).

CAS PubMed Google Scholar

Kukita, Y. et al. Selective identification of somatic mutations in pancreatic cancer cells through a combination of next-generation sequencing of plasma DNA using molecular barcodes and a bioinformatic variant filter. PLoS ONE 13, e0192611 (2018).

PubMed PubMed Central Google Scholar

Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 17541760 (2009).

CAS PubMed PubMed Central Google Scholar

Koboldt, D. C. et al. VarScan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 22, 568576 (2012).

CAS PubMed PubMed Central Google Scholar

Kukita, Y. et al. Quantitative identification of mutant alleles derived from lung cancer in plasma cell-free DNA via anomaly detection using deep sequencing data. PLoS ONE 8, e81468 (2013).

ADS PubMed PubMed Central Google Scholar

Cao, Y., Xie, L., Xie, Y. & Xu, H. Sequential change-point detection via online convex optimization. Entropy 20, 108 (2018).

ADS PubMed Central Google Scholar

Link:
Fragment size and dynamics of EGFR-mutated tumor-derived DNA provide prognostic information regarding EGFR-TKI efficacy in patients with EGFR-mutated...

Scientists Unlock the Secrets of Cellular Aging: What Happens After You Turn 70? – SciTechDaily

Researchers have revealed the cellular mysteries behind aging.

A new explanation for aging has been developed by researchers who have shown that genetic abnormalities that develop gradually over a lifetime cause substantial alterations in how blood is generated beyond the age of 70.

According to recent research, the drastic reduction in blood production beyond the age of 70 is likely caused by genetic alterations that steadily accumulate in blood stem cells throughout life.

Researchers from the Wellcome Sanger Institute, the Wellcome-MRC Cambridge Stem Cell Institute, and others have published a study that offers a new theory of aging in the journal Nature.

Somatic mutations, or alterations to the genetic code, occur in all human cells during the course of a lifetime. Aging is most likely caused by the accumulation of numerous sorts of damage to our cells over time, with one hypothesis proposing that the accumulation of somatic mutations causes cells to gradually lose functional reserve. However, it is still unknown how such slow-building molecular damage may result in the rapid decline in organ performance around the age of 70.

The Wellcome Sanger Institute, the Cambridge Stem Cell Institute, and collaborators examined the production of blood cells from the bone marrow in 10 people ranging in age from newborns to the elderly in order to better understand how the body ages.

3,579 blood stem cells had their whole genomes sequenced, allowing researchers to determine every somatic mutation present in each cell. Using this information, the team was able to create family trees of each persons blood stem cells, providing for the first time an impartial perspective of the connections between blood cells and how these ties develop over the course of a persons lifetime.

After the age of 70 years, the researchers discovered that these family trees underwent significant change. In adults under the age of 65, 20,000 to 200,000 stem cells contributed roughly equal amounts to the creation of blood cells. In contrast, blood production was exceedingly uneven in those above the age of 70.

In every elderly person investigated, a small number of enlarged stem cell clonesas few as 10 to 20contributed as much as half of the total blood output. Because of an uncommon class of somatic mutations known as driver mutations, these highly active stem cells have gradually increased in number during that persons life.

These findings led the team to propose a model in which age-associated changes in blood production come from somatic mutations causing selfish stem cells to dominate the bone marrow in the elderly. This model, with the steady introduction of driver mutations that cause the growth of functionally altered clones over decades, explains the dramatic and inevitable shift to reduced diversity of blood cell populations after the age of 70. Which clones become dominant varies from person to person, and so the model also explains the variation seen in disease risk and other characteristics in older adults. A second study, also published in Nature, explores how different individual driver mutations affect cell growth rates over time.

Dr. Emily Mitchell, Haematology Registrar at Addenbrookes Hospital, a Ph.D. student at the Wellcome Sanger Institute, and lead researcher on the study, said: Our findings show that the diversity of blood stem cells is lost in older age due to positive selection of faster-growing clones with driver mutations.

These clones outcompete the slower-growing ones. In many cases this increased fitness at the stem cell level likely comes at a cost their ability to produce functional mature blood cells is impaired, so explaining the observed age-related loss of function in the blood system.

Dr. Elisa Laurenti, Assistant Professor and Wellcome Royal Society Sir Henry Dale Fellow at the Wellcome-MRC Cambridge Stem Cell Institute at the University of Cambridge, and joint senior researcher on this study, said: Factors such as chronic inflammation, smoking, infection, and chemotherapy cause earlier growth of clones with cancer-driving mutations. We predict that these factors also bring forward the decline in blood stem cell diversity associated with aging. It is possible that there are factors that might slow this process down, too. We now have the exciting task of figuring out how these newly discovered mutations affect blood function in the elderly, so we can learn how to minimize disease risk and promote healthy aging.

Dr. Peter Campbell, Head of the Cancer, Ageing and Somatic Mutation Programme at the Wellcome Sanger Institute, and senior researcher on the study said: Weve shown, for the first time, how steadily accumulating mutations throughout life lead to a catastrophic and inevitable change in blood cell populations after the age of 70. What is super exciting about this model is that it may well apply to other organ systems too. We see these selfish clones with driver mutations expanding with age in many other tissues of the body we know this can increase cancer risk, but it could also be contributing to other functional changes associated with aging.

References: Clonal dynamics of haematopoiesis across the human lifespan by Emily Mitchell, Michael Spencer Chapman, Nicholas Williams, Kevin J. Dawson, Nicole Mende, Emily F. Calderbank, Hyunchul Jung, Thomas Mitchell, Tim H. H. Coorens, David H. Spencer, Heather Machado, Henry Lee-Six, Megan Davies, Daniel Hayler, Margarete A. Fabre, Krishnaa Mahbubani, Federico Abascal, Alex Cagan, George S. Vassiliou, Joanna Baxter, Inigo Martincorena, Michael R. Stratton, David G. Kent, Krishna Chatterjee, Kourosh Saeb Parsy, Anthony R. Green, Jyoti Nangalia, Elisa Laurenti, and Peter J. Campbell, 1 June 2022, Nature. DOI: 10.1038/s41586-022-04786-y

The longitudinal dynamics and natural history of clonal haematopoiesis by Margarete A. Fabre, Jos Guilherme de Almeida, Edoardo Fiorillo, Emily Mitchell, Aristi Damaskou, Justyna Rak, Valeria Orr, Michele Marongiu, Michael Spencer Chapman, M. S. Vijayabaskar, Joanna Baxter, Claire Hardy, Federico Abascal, Nicholas Williams, Jyoti Nangalia, Iigo Martincorena, Peter J. Campbell, Eoin F. McKinney, Francesco Cucca, Moritz Gerstung, and George S. Vassiliou, 1 June 2022, Nature. DOI: 10.1038/s41586-022-04785-z

The study was funded by Wellcome and the William B Harrison Foundation.

Excerpt from:
Scientists Unlock the Secrets of Cellular Aging: What Happens After You Turn 70? - SciTechDaily

Researchers revive abandoned technique in effort to make artificial human eggs in a test tube – STAT

In a little-noticed study published earlier this year, scientists from Oregon Health & Science University reported the birth of three mouse pups that had been created with a never-before-used recipe for reproduction. Using a common cloning technique, researchers removed the genetic material from one females eggs and replaced them with nuclear DNA from the skin cells of another. Then with a novel chemical cocktail, they nudged the eggs to lose half their new sets of chromosomes and fertilized them with mouse sperm.

In a big step toward achieving in vitro gametogenesis one of reproductive medicines more ambitious moonshots the group led by pioneering fertility researcher Shoukrat Mitalipov now intends to use the same method to make artificial human embryos in a test tube.

If successful, the research holds enormous potential for treating infertility, preventing heritable diseases, and opening up the possibility for same-sex couples to have genetically related children.

advertisement

Its one of those high-risk, high reward type of projects, said Paula Amato, an OB-GYN and infertility specialist at OHSU who collects the human eggs used in Mitalipovs experiments. We have no idea yet if it will work, but age-related fertility decline remains an intractable problem in our field, so were eternally grateful to these private funders who are filling a real need here.

Mitalipov directs the Center for Embryonic Cell and Gene Therapy at OHSU. Established in 2013, the center focuses on combining assisted reproductive technologies with genetic correction techniques, with the goal of one day preventing inherited disease.

advertisement

Related:

The groups work on in vitro gametogenesis (IVG) in human cells is being made possible by an award from Open Philanthropy a grant-making organization primarily funded by Facebook co-founder Dustin Moskovitz and his wife Cari Tuna which will supply the researchers with $4 million over the next three years. The infusion of funds and the involvement of a scientist as storied as Mitalipov makes the ethical and legal questions surrounding mass egg and sperm production more urgent, experts told STAT.

In the U.S., there are no federal laws that prohibit this type of IVG work. However, Congress has barred any research that creates, destroys, or knowingly harms human embryos from receiving federal funding. At the state level, laws governing human embryo research vary widely with 11 states banning it entirely, five states expressly permitting it, and a lot of gray areas in between.

For IVG to move from the research lab to a fertility clinic would require permission from the Food and Drug Administration. Its still unclear if thats something the agency would be able to consider a spending bill rider currently prevents the FDA from receiving any requests to pursue clinical trials involving starting pregnancies with embryos that have been genetically manipulated. In 2019, Congress considered modifying the ban, following a push from scientists and advocates of mitochondrial replacement therapy, also known as three-person IVF, but ultimately renewed it. Mitochondrial replacement therapy is a procedure that combines genetic material from an egg and sperm with mitochondria from a female donor.

Somatic cell nuclear transfer for IVG could fall under the same provision, if the somatic DNA and the egg came from different people. But if they came from the same person, that might represent a loophole.

Related:

Some bioethicists worry that the easy availability of IVG could usher in a new era of eugenics, scenarios where prospective parents could create large numbers of embryos and use genetic tools to select the best one. IVG also raises the specter of nonconsensual parenthood something most state laws are currently ill-equipped to handle.

Should this become clinically available, there will be legitimate questions about whose cells can be used and under what conditions that will need regulatory answers, said Hank Greely, director of the Stanford Center for Law and Bioscience, whose book, The End of Sex, examines the future of in vitro gametogenesis. Will that happen? We dont know. But Mitalipov has certainly proven himself a bold and creative scientist, and from my perspective, having his group join the effort to help people who want to have genetic babies but cant is a good thing, provided they can do it safely and effectively.

Mitalipovs lab has long been an incubator for envelope-pushing science. In 2009, he and his colleagues figured out a way to swap out glitchy mitochondrial DNA for healthy versions in the egg cells of monkeys a groundbreaking advance that paved the way for mitochondrial replacement therapy in humans. In 2013, they created lines of embryonic stem cells from cloned human embryos for the first time. A few years later, they became the first team in the U.S. to attempt to correct a genetic mutation in viable human embryos using CRISPR.

But until recently, in vitro gametogenesis, or IVG, wasnt on his to-do list.

Gametes are the cells capable of giving rise to future generations: sperm and eggs. The idea behind IVG is to produce those kinds of cells in test tubes, rather than inside a developing animals body.

In recent years, scientists have made headlines producing artificial gametes from induced pluripotent stem cells. But Mitalipovs group plans to revive a much older technology, which saw some early success in IVG before being abandoned: somatic cell nuclear transfer.

Somatic cell nuclear transfer was pioneered by researchers at the Roslin Institute in Scotland. After they succeeded in using the technique to clone the first mammal a sheep named Dolly scientists realized it might be used to generate artificial gametes, if they could overcome a few additional hurdles.

In cloning, the emptied egg receives a full set of chromosomes from the somatic cell donor and is stimulated in the lab to make it start dividing. Any offspring that result will be genetically identical to that somatic cell.

The procedure for making an artificial oocyte is technically similar to cloning, but would generate unique individuals after fertilization with sperm. However, in order for any resulting embryos to have the right number of chromosomes, the donor DNA has to be cut in half, a process known as haploidization. Oocytes are equipped with the machinery to make that adjustment, if the somatic DNA is introduced at the right phase of their cell cycle.

In 2000, four years after Dolly was born, researchers in Spain generated the first human artificial oocytes using this method. They fertilized three of them, and froze the resulting embryos at the two-cell stage. The plan was to transfer the frozen embryos to the uterus of a woman who had been unable to conceive, and consented to having her somatic DNA slipped into donor eggs as a last-ditch attempt to have genetically related children with her husband.

Related:

But when the same protocol was tested in mice where its effects could be examined more closely the chromosomes didnt separate as intended. Shortly thereafter, somatic cell nuclear transfer for human reproduction was banned in many countries, including Spain.

The IVG field moved on, buoyed by the discovery a few years later of a method for taking any kind of cell and rewinding its developmental clock to a more primitive state. With the right chemical cues, a team of Japanese scientists nudged these pluripotent stem cells to produce functional gametes in mice; first sperm in 2011, then eggs, five years later. But they struggled to generate similar results in humans.

In 2018, the group succeeded for the first time in making immature human eggs from scratch. But the process wasnt very efficient and it involved incubating the human stem cells in mini-ovaries theyd created in the lab from mouse embryonic cells a resource-intensive process not exactly suited to mass manufacturing.

So when a post-doc at OHSU named Eunju Kang proposed revisiting the idea of somatic cell nuclear transfer for IVG, Mitalipov was initially skeptical. But data from her initial mouse experiments proved persuasive. Mitalipov threw his support behind the project, and teamed up with a group at Weill Cornell Medicine in New York, including reproductive endocrinologist Gianpiero Palermo, who had successfully generated artificial human oocytes using cloning technology back in 2002. They published the results of their mice experiments in Nature Communications Biology in January.

Related:

The OHSU team is now adapting those methods to see if they can generate artificial human eggs with properly separated chromosomes. If successful, they plan to then fertilize those eggs with sperm and grow the resulting embryos in the lab for five or six days to see if they develop normally.

They are betting that this method, while older, will prove better than the induced pluripotent stem cell technologies currently being advanced by artificial egg-making start-up outfits like Conception, Ivy Natal and Gameto.

That approach requires the cells to be cultured for months rather than days, which can lead to epigenetic programming errors and chromosomal instability. Mitalipov also believes that starting with natural eggs will make it easier to strip the donor DNA of its cellular memory and return it to the primitive state known as totipotency a critical step in enabling the embryo to eventually develop all the specialized tissues that make up a human body.

Science Writer

Megan Molteni is a science writer for STAT, covering genomic medicine, neuroscience, and reproductive tech.

This name will appear with your comment

Link:
Researchers revive abandoned technique in effort to make artificial human eggs in a test tube - STAT

Genome-wide analyses of 200,453 individuals yield new insights into the causes and consequences of clonal hematopoiesis – Nature.com

Zhang, L. & Vijg, J. Somatic mutagenesis in mammals and its implications for human disease and aging. Annu. Rev. Genet. 52, 397419 (2018).

CAS PubMed PubMed Central Article Google Scholar

Martincorena, I. & Campbell, P. J. Somatic mutation in cancer and normal cells. Science 349, 14831489 (2015).

CAS PubMed Article Google Scholar

Kakiuchi, N. & Ogawa, S. Clonal expansion in non-cancer tissues. Nat. Rev. Cancer 21, 239256 (2021).

CAS PubMed Article Google Scholar

Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 24772487 (2014).

PubMed PubMed Central Article CAS Google Scholar

Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 24882498 (2014).

PubMed PubMed Central Article CAS Google Scholar

McKerrell, T. et al. Leukemia-associated somatic mutations drive distinct patterns of age-related clonal hemopoiesis. Cell Rep. 10, 12391245 (2015).

CAS PubMed PubMed Central Article Google Scholar

Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 14721478 (2014).

CAS PubMed PubMed Central Article Google Scholar

Abelson, S. et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature 559, 400404 (2018).

CAS PubMed PubMed Central Article Google Scholar

Desai, P. et al. Somatic mutations precede acute myeloid leukemia years before diagnosis. Nat. Med. 24, 10151023 (2018).

CAS PubMed PubMed Central Article Google Scholar

Jaiswal, S. et al. Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N. Engl. J. Med. 377, 111121 (2017).

PubMed PubMed Central Article Google Scholar

Dorsheimer, L. et al. Association of mutations contributing to clonal hematopoiesis with prognosis in chronic ischemic heart failure. JAMA Cardiol. 4, 2533 (2019).

PubMed Article Google Scholar

Jaiswal, S. & Ebert, B. L. Clonal hematopoiesis in human aging and disease. Science 366, eaan4673 (2019).

CAS PubMed PubMed Central Article Google Scholar

Coombs, C. C. et al. Therapy-related clonal hematopoiesis in patients with non-hematologic cancers is common and associated with adverse clinical outcomes. Cell Stem Cell 21, 374382.e4 (2017).

CAS PubMed PubMed Central Article Google Scholar

Gibson, C. J. et al. Clonal hematopoiesis associated with adverse outcomes after autologous stem-cell transplantation for lymphoma. J. Clin. Oncol. 35, 15981605 (2017).

CAS PubMed PubMed Central Article Google Scholar

Meisel, M. et al. Microbial signals drive pre-leukaemic myeloproliferation in a Tet2-deficient host. Nature 557, 580584 (2018).

CAS PubMed PubMed Central Article Google Scholar

Yoshizato, T. et al. Somatic mutations and clonal hematopoiesis in aplastic anemia. N. Engl. J. Med. 373, 3547 (2015).

CAS PubMed PubMed Central Article Google Scholar

Bick, A. G. et al. Inherited causes of clonal haematopoiesis in 97,691 whole genomes. Nature 586, 763768 (2020).

CAS PubMed PubMed Central Article Google Scholar

Hinds, D. A. et al. Germ line variants predispose to both JAK2 V617F clonal hematopoiesis and myeloproliferative neoplasms. Blood 128, 11211128 (2016).

CAS PubMed PubMed Central Article Google Scholar

Szustakowski, J. D. et al. Advancing human genetics research and drug discovery through exome sequencing of the UK Biobank. Nat. Genet. 53, 942948 (2021).

CAS PubMed Article Google Scholar

Fabre, M. A. et al. The longitudinal dynamics and natural history of clonal haematopoiesis. Nature 606, 335342 (2022).

CAS PubMed PubMed Central Article Google Scholar

De-Morgan, A., Meggendorfer, M., Haferlach, C. & Shlush, L. Male predominance in AML is associated with specific preleukemic mutations. Leukemia 35, 867870 (2021).

PubMed Article Google Scholar

Bick, A. G. et al. Genetic interleukin 6 signaling deficiency attenuates cardiovascular risk in clonal hematopoiesis. Circulation 141, 124131 (2020).

CAS PubMed Article Google Scholar

Bulik-Sullivan, B. K. et al. LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291295 (2015).

CAS PubMed PubMed Central Article Google Scholar

Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 12281235 (2015).

CAS PubMed PubMed Central Article Google Scholar

Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 11931203 (2016).

CAS PubMed PubMed Central Article Google Scholar

Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621629 (2018).

CAS PubMed PubMed Central Article Google Scholar

Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369375 (2012) .

CAS PubMed PubMed Central Article Google Scholar

Walsh, T. G. et al. Loss of the exocyst complex component EXOC3 promotes hemostasis and accelerates arterial thrombosis. Blood Adv. 5, 674686 (2021).

CAS PubMed PubMed Central Article Google Scholar

Astle, W. J. et al. The allelic landscape of human blood cell trait variation and links to common complex disease. Cell 167, 14151429.e19 (2016).

CAS PubMed PubMed Central Article Google Scholar

Ning, Z., Pawitan, Y. & Shen, X. High-definition likelihood inference of genetic correlations across human complex traits. Nat. Genet. 52, 859864 (2020).

CAS PubMed Article Google Scholar

Zekavat, S. M. et al. Hematopoietic mosaic chromosomal alterations increase the risk for diverse types of infection. Nat. Med. 27, 10121024 (2021).

CAS PubMed PubMed Central Article Google Scholar

Niroula, A. et al. Distinction of lymphoid and myeloid clonal hematopoiesis. Nat. Med. 27, 19211927 (2021).

CAS PubMed PubMed Central Article Google Scholar

Kamat, M. A. et al. PhenoScanner V2: an expanded tool for searching human genotype-phenotype associations. Bioinformatics 35, 48514853 (2019).

CAS PubMed PubMed Central Article Google Scholar

Staley, J. R. et al. PhenoScanner: a database of human genotype-phenotype associations. Bioinformatics 32, 32073209 (2016).

CAS PubMed PubMed Central Article Google Scholar

Vsa, U. et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat. Genet. 53, 13001310 (2021).

PubMed PubMed Central Article CAS Google Scholar

Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481487 (2016).

CAS PubMed Article Google Scholar

Ragu, C. et al. The transcription factor Srf regulates hematopoietic stem cell adhesion. Blood 116, 44644473 (2010).

CAS PubMed Article Google Scholar

Zink, F. et al. Clonal hematopoiesis, with and without candidate driver mutations, is common in the elderly. Blood 130, 742752 (2017).

CAS PubMed PubMed Central Article Google Scholar

Zhou, G. et al. NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis. Nucleic Acids Res. 47, W234W241 (2019).

CAS PubMed PubMed Central Article Google Scholar

Cowen, L., Ideker, T., Raphael, B. J. & Sharan, R. Network propagation: a universal amplifier of genetic associations. Nat. Rev. Genet. 18, 551562 (2017).

CAS PubMed Article Google Scholar

Fernndez-Tajes, J. et al. Developing a network view of type 2 diabetes risk pathways through integration of genetic, genomic and functional data. Genome Med. 11, 19 (2019).

PubMed PubMed Central Article Google Scholar

Tischkowitz, M. et al. Bi-allelic silencing of the Fanconi anaemia gene FANCF in acute myeloid leukaemia. Br. J. Haematol. 123, 469471 (2003).

CAS PubMed Article Google Scholar

Lim, Y. et al. Integration of Hedgehog and mutant FLT3 signaling in myeloid leukemia. Sci. Transl. Med. 7, 291ra96 (2015).

PubMed PubMed Central Article CAS Google Scholar

Ostrander, E. L. et al. The GNASR201C mutation associated with clonal hematopoiesis supports transplantable hematopoietic stem cell activity. Exp. Hematol. 57, 1420 (2018).

CAS PubMed Article Google Scholar

Ghoussaini, M. et al. Open Targets Genetics: systematic identification of trait-associated genes using large-scale genetics and functional genomics. Nucleic Acids Res. 49, D1311D1320 (2021).

CAS PubMed Article Google Scholar

Taylor, K. E., Ansel, K. M., Marson, A., Criswell, L. A. & Farh, K. K.-H. PICS2: next-generation fine mapping via probabilistic identification of causal SNPs. Bioinformatics 37, 30043007 (2021).

CAS PubMed Central Article Google Scholar

Farh, K. K.-H. et al. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337343 (2015).

CAS PubMed Article Google Scholar

Bao, E. L. et al. Inherited myeloproliferative neoplasm risk affects haematopoietic stem cells. Nature 586, 769775 (2020).

CAS PubMed PubMed Central Article Google Scholar

Buenrostro, J. D. et al. Integrated single-cell analysis maps the continuous regulatory landscape of human hematopoietic differentiation. Cell 173, 15351548.e16 (2018).

CAS PubMed PubMed Central Article Google Scholar

Ulirsch, J. C. et al. Interrogation of human hematopoiesis at single-cell and single-variant resolution. Nat. Genet. 51, 683693 (2019).

CAS PubMed PubMed Central Article Google Scholar

See the original post here:
Genome-wide analyses of 200,453 individuals yield new insights into the causes and consequences of clonal hematopoiesis - Nature.com

Escargot controls somatic stem cell maintenance through the attenuation of the insulin receptor pathway in Drosophila – Newswise

Adultstem cellscoordinate intrinsic and extrinsic, local and systemic, cues to maintain the proper balance between self-renewal and differentiation. However, the precise mechanisms stem cells use to integrate these signals remain elusive. Here, we show that Escargot (Esg), a member of the Snail family of transcription factors, regulates the maintenance of somatic cyst stem cells (CySCs) in theDrosophilatestisby attenuating the activity of the pro-differentiationinsulin receptor(InR) pathway. Esg positively regulates the expression of an antagonist ofinsulin signaling,ImpL2, while also attenuating the expression ofInR. Furthermore, Esg-mediated repression of the InR pathway is required to suppress CySC loss in response to starvation. Given the conservation of Snail-family transcription factors, characterizing the mechanisms by which Esg regulates cell-fate decisions duringhomeostasisand a decline in nutrient availability is likely to provide insight into themetabolic regulationof stem cell behavior in other tissues and organisms.

Here is the original post:
Escargot controls somatic stem cell maintenance through the attenuation of the insulin receptor pathway in Drosophila - Newswise