fbpx
Wikipedia

Hematopoietic stem cell

Hematopoietic stem cells (HSCs) are the stem cells[1] that give rise to other blood cells. This process is called haematopoiesis.[2] In vertebrates, the first definitive HSCs arise from the ventral endothelial wall of the embryonic aorta within the (midgestational) aorta-gonad-mesonephros region, through a process known as endothelial-to-hematopoietic transition.[3][4] In adults, haematopoiesis occurs in the red bone marrow, in the core of most bones. The red bone marrow is derived from the layer of the embryo called the mesoderm.

Hematopoietic stem cell
Overview of normal human haematopoiesis
Details
PrecursorHemangioblast
SystemHematopoietic system
LocationBone marrow
FunctionStem cells that give rise to other blood cells
Identifiers
Latincellula haematopoietica praecursoria
Acronym(s)HSC
MeSHD006412
THH2.00.01.0.00006
Anatomical terms of microanatomy
[edit on Wikidata]

Haematopoiesis is the process by which all mature blood cells are produced. It must balance enormous production needs (the average person produces more than 500 billion blood cells every day) with the need to regulate the number of each blood cell type in the circulation. In vertebrates, the vast majority of hematopoiesis occurs in the bone marrow and is derived from a limited number of hematopoietic stem cells that are multipotent and capable of extensive self-renewal.

Hematopoietic stem cells give rise to different types of blood cells, in lines called myeloid and lymphoid. Myeloid and lymphoid lineages both are involved in dendritic cell formation. Myeloid cells include monocytes, macrophages, neutrophils, basophils, eosinophils, erythrocytes, and megakaryocytes to platelets. Lymphoid cells include T cells, B cells, natural killer cells, and innate lymphoid cells.

The definition of hematopoietic stem cell has developed since they were first discovered in 1961.[5] The hematopoietic tissue contains cells with long-term and short-term regeneration capacities and committed multipotent, oligopotent, and unipotent progenitors. Hematopoietic stem cells constitute 1:10,000 of cells in myeloid tissue.

HSC transplants are used in the treatment of cancers and other immune system disorders[6] due to their regenerative properties. [7]

Structure edit

They are round, non-adherent, with a rounded nucleus and low cytoplasm-to-nucleus ratio. In shape, hematopoietic stem cells resemble lymphocytes.

Location edit

The very first hematopoietic stem cells during (mouse and human) embryonic development are found in aorta-gonad-mesonephros region and the vitelline and umbilical arteries.[8][9][10] Slightly later, HSCs are also found in the placenta, yolk sac, embryonic head, and fetal liver.[3][11]

Stem and progenitor cells can be taken from the pelvis, at the iliac crest, using a needle and syringe.[12] The cells can be removed as liquid (to perform a smear to look at the cell morphology) or they can be removed via a core biopsy (to maintain the architecture or relationship of the cells to each other and to the bone).[citation needed]

Subtypes edit

A colony-forming unit is a subtype of HSC. (This sense of the term is different from colony-forming units of microbes, which is a cell counting unit.) There are various kinds of HSC colony-forming units:

The above CFUs are based on the lineage. Another CFU, the colony-forming unit–spleen (CFU-S), was the basis of an in vivo clonal colony formation, which depends on the ability of infused bone marrow cells to give rise to clones of maturing hematopoietic cells in the spleens of irradiated mice after 8 to 12 days. It was used extensively in early studies, but is now considered to measure more mature progenitor or transit-amplifying cells rather than stem cells[citation needed].

Isolating stem cells edit

Since hematopoietic stem cells cannot be isolated as a pure population, it is not possible to identify them in a microscope.[citation needed] Hematopoietic stem cells can be identified or isolated by the use of flow cytometry where the combination of several different cell surface markers (particularly CD34) are used to separate the rare hematopoietic stem cells from the surrounding blood cells. Hematopoietic stem cells lack expression of mature blood cell markers and are thus called Lin-. Lack of expression of lineage markers is used in combination with detection of several positive cell-surface markers to isolate hematopoietic stem cells. In addition, hematopoietic stem cells are characterised by their small size and low staining with vital dyes such as rhodamine 123 (rhodamine lo) or Hoechst 33342 (side population).

Function edit

 
Diagram of cells that arise from Hematopoietic stem cells during the process of hematopoiesis.

Haematopoiesis edit

Hematopoietic stem cells are essential to haematopoiesis, the formation of the cells within blood. Hematopoietic stem cells can replenish all blood cell types (i.e., are multipotent) and self-renew. A small number of hematopoietic stem cells can expand to generate a very large number of daughter hematopoietic stem cells. This phenomenon is used in bone marrow transplantation,[13] when a small number of hematopoietic stem cells reconstitute the hematopoietic system. This process indicates that, subsequent to bone marrow transplantation, symmetrical cell divisions into two daughter hematopoietic stem cells must occur.

Stem cell self-renewal is thought to occur in the stem cell niche in the bone marrow, and it is reasonable to assume that key signals present in this niche will be important in self-renewal.[2] There is much interest in the environmental and molecular requirements for HSC self-renewal, as understanding the ability of HSC to replenish themselves will eventually allow the generation of expanded populations of HSC in vitro that can be used therapeutically.

Quiescence edit

Hematopoietic stem cells, like all adult stem cells, mostly exist in a state of quiescence, or reversible growth arrest. The altered metabolism of quiescent HSCs helps the cells survive for extended periods of time in the hypoxic bone marrow environment.[14] When provoked by cell death or damage, Hematopoietic stem cells exit quiescence and begin actively dividing again. The transition from dormancy to propagation and back is regulated by the MEK/ERK pathway and PI3K/AKT/mTOR pathway.[15] Dysregulation of these transitions can lead to stem cell exhaustion, or the gradual loss of active Hematopoietic stem cells in the blood system.[15]

Mobility edit

Hematopoietic stem cells have a higher potential than other immature blood cells to pass the bone marrow barrier, and, thus, may travel in the blood from the bone marrow in one bone to another bone. If they settle in the thymus, they may develop into T cells. In the case of fetuses and other extramedullary hematopoiesis. Hematopoietic stem cells may also settle in the liver or spleen and develop.

This enables Hematopoietic stem cells to be harvested directly from the blood.

Clinical significance edit

Transplant edit

Hematopoietic stem cell transplantation (HSCT) is the transplantation of multipotent hematopoietic stem cells, usually derived from bone marrow, peripheral blood, or umbilical cord blood.[16][17][18] It may be autologous (the patient's own stem cells are used), allogeneic (the stem cells come from a donor) or syngeneic (from an identical twin).[16][17]

It is most often performed for patients with certain cancers of the blood or bone marrow, such as multiple myeloma or leukemia.[17] In these cases, the recipient's immune system is usually destroyed with radiation or chemotherapy before the transplantation. Infection and graft-versus-host disease are major complications of allogeneic HSCT.[17]

In order to harvest stem cells from the circulating peripheral blood, blood donors are injected with a cytokine, such as granulocyte-colony stimulating factor (G-CSF), that induces cells to leave the bone marrow and circulate in the blood vessels.[19] In mammalian embryology, the first definitive Hematopoietic stem cells are detected in the AGM (aorta-gonad-mesonephros), and then massively expanded in the fetal liver prior to colonising the bone marrow before birth.[20]

Hematopoietic stem cell transplantation remains a dangerous procedure with many possible complications; it is reserved for patients with life-threatening diseases. As survival following the procedure has increased, its use has expanded beyond cancer to autoimmune diseases[21][22] and hereditary skeletal dysplasias; notably malignant infantile osteopetrosis[23][24] and mucopolysaccharidosis.[25]

Stem cells can be used to regenerate different types of tissues. HCT is an established as therapy for chronic myeloid leukemia, acute lymphatic leukemia, aplastic anemia, and hemoglobinopathies, in addition to acute myeloid leukemia and primary immune deficiencies. Hematopoietic system regeneration is typically achieved within 2-4 weeks post-chemo- or irradiation therapy and HCT. HSCs are being clinically tested for their use in non-hematopoietic tissue regeneration.[26]

Aging of hematopoietic stem cells edit

DNA damage edit

DNA strand breaks accumulate in long term hematopoietic stem cells during aging.[27] This accumulation is associated with a broad attenuation of DNA repair and response pathways that depends on HSC quiescence.[27] Non-homologous end joining (NHEJ) is a pathway that repairs double-strand breaks in DNA. NHEJ is referred to as "non-homologous" because the break ends are directly ligated without the need for a homologous template. The NHEJ pathway depends on several proteins including ligase 4, DNA polymerase mu and NHEJ factor 1 (NHEJ1, also known as Cernunnos or XLF).

DNA ligase 4 (Lig4) has a highly specific role in the repair of double-strand breaks by NHEJ. Lig4 deficiency in the mouse causes a progressive loss of hematopoietic stem cells during aging.[28] Deficiency of lig4 in pluripotent stem cells results in accumulation of DNA double-strand breaks and enhanced apoptosis.[29]

In polymerase mu mutant mice, hematopoietic cell development is defective in several peripheral and bone marrow cell populations with about a 40% decrease in bone marrow cell number that includes several hematopoietic lineages.[30] Expansion potential of hematopoietic progenitor cells is also reduced. These characteristics correlate with reduced ability to repair double-strand breaks in hematopoietic tissue.

Deficiency of NHEJ factor 1 in mice leads to premature aging of hematopoietic stem cells as indicated by several lines of evidence including evidence that long-term repopulation is defective and worsens over time.[31] Using a human induced pluripotent stem cell model of NHEJ1 deficiency, it was shown that NHEJ1 has an important role in promoting survival of the primitive hematopoietic progenitors.[32] These NHEJ1 deficient cells possess a weak NHEJ1-mediated repair capacity that is apparently incapable of coping with DNA damages induced by physiological stress, normal metabolism, and ionizing radiation.[32]

The sensitivity of hematopoietic stem cells to Lig4, DNA polymerase mu and NHEJ1 deficiency suggests that NHEJ is a key determinant of the ability of stem cells to maintain themselves against physiological stress over time.[28] Rossi et al.[33] found that endogenous DNA damage accumulates with age even in wild type Hematopoietic stem cells, and suggested that DNA damage accrual may be an important physiological mechanism of stem cell aging.

Loss of clonal diversity edit

A study shows the clonal diversity of hematopoietic stem cells gets drastically reduced around age 70 to a faster-growing few, substantiating a novel theory of ageing which could enable healthy aging.[34][35] Of note, the shift in clonal diversity during aging was previously reported in 2008[36] for the murine system by the Christa Muller-Sieburg laboratory in San Diego, California.

Research edit

Behavior in culture edit

A cobblestone area-forming cell (CAFC) assay is a cell culture-based empirical assay. When plated onto a confluent culture of stromal feeder layer,[37] a fraction of hematopoietic stem cells creep between the gaps (even though the stromal cells are touching each other) and eventually settle between the stromal cells and the substratum (here the dish surface) or trapped in the cellular processes between the stromal cells. Emperipolesis is the in vivo phenomenon in which one cell is completely engulfed into another (e.g. thymocytes into thymic nurse cells); on the other hand, when in vitro, lymphoid lineage cells creep beneath nurse-like cells, the process is called pseudoemperipolesis. This similar phenomenon is more commonly known in the HSC field by the cell culture terminology cobble stone area-forming cells (CAFC), which means areas or clusters of cells look dull cobblestone-like under phase contrast microscopy, compared to the other hematopoietic stem cells, which are refractile. This happens because the cells that are floating loosely on top of the stromal cells are spherical and thus refractile. However, the cells that creep beneath the stromal cells are flattened and, thus, not refractile. The mechanism of pseudoemperipolesis is only recently coming to light. It may be mediated by interaction through CXCR4 (CD184) the receptor for CXC Chemokines (e.g., SDF1) and α4β1 integrins.[38]

Repopulation kinetics edit

Hematopoietic stem cells (HSC) cannot be easily observed directly, and, therefore, their behaviors need to be inferred indirectly. Clonal studies are likely the closest technique for single cell in vivo studies of HSC. Here, sophisticated experimental and statistical methods are used to ascertain that, with a high probability, a single HSC is contained in a transplant administered to a lethally irradiated host. The clonal expansion of this stem cell can then be observed over time by monitoring the percent donor-type cells in blood as the host is reconstituted. The resulting time series is defined as the repopulation kinetic of the HSC.

The reconstitution kinetics are very heterogeneous. However, using symbolic dynamics, one can show that they fall into a limited number of classes.[39] To prove this, several hundred experimental repopulation kinetics from clonal Thy-1lo SCA-1+ lin(B220, CD4, CD8, Gr-1, Mac-1 and Ter-119)[40] c-kit+ HSC were translated into symbolic sequences by assigning the symbols "+", "-", "~" whenever two successive measurements of the percent donor-type cells have a positive, negative, or unchanged slope, respectively. By using the Hamming distance, the repopulation patterns were subjected to cluster analysis yielding 16 distinct groups of kinetics. To finish the empirical proof, the Laplace add-one approach was used to determine that the probability of finding kinetics not contained in these 16 groups is very small. By corollary, this result shows that the hematopoietic stem cell compartment is also heterogeneous by dynamical criteria.

It was originally believed that all hematopoietic stem cells were alike in their self-renewal and differentiation abilities. This view was first challenged by the 2002 discovery by the Muller-Sieburg group in San Diego, who illustrated that different stem cells can show distinct repopulation patterns that are epigenetically predetermined intrinsic properties of clonal Thy-1lo Sca-1+ lin c-kit+ HSC.[41][42][43] The results of these clonal studies led to the notion of lineage bias. Using the ratio   of lymphoid (L) to myeloid (M) cells in blood as a quantitative marker, the stem cell compartment can be split into three categories of HSC. Balanced (Bala) hematopoietic stem cells repopulate peripheral white blood cells in the same ratio of myeloid to lymphoid cells as seen in unmanipulated mice (on average about 15% myeloid and 85% lymphoid cells, or 3 ≤ ρ ≤ 10). Myeloid-biased (My-bi) hematopoietic stem cells give rise to very few lymphocytes resulting in ratios 0 < ρ < 3, while lymphoid-biased (Ly-bi) hematopoietic stem cells generate very few myeloid cells, which results in lymphoid-to-myeloid ratios of ρ > 10. All three types are normal types of HSC, and they do not represent stages of differentiation. Rather, these are three classes of HSC, each with an epigenetically fixed differentiation program. These studies also showed that lineage bias is not stochastically regulated or dependent on differences in environmental influence. My-bi HSC self-renew longer than balanced or Ly-bi HSC. The myeloid bias results from reduced responsiveness to the lymphopoetin interleukin 7 (IL-7).[42]

Subsequently, other groups confirmed and highlighted the original findings.[44] For example, the Eaves group confirmed in 2007 that repopulation kinetics, long-term self-renewal capacity, and My-bi and Ly-bi are stably inherited intrinsic HSC properties.[45] In 2010, the Goodell group provided additional insights about the molecular basis of lineage bias in side population (SP) SCA-1+ lin c-kit+ HSC.[46] As previously shown for IL-7 signaling, it was found that a member of the transforming growth factor family (TGF-beta) induces and inhibits the proliferation of My-bi and Ly-bi HSC, respectively.

Etymology edit

From Greek haimato-, combining form of haima 'blood', and from the Latinized form of Greek poietikos 'capable of making, creative, productive', from poiein 'to make, create'.[47]

See also edit

References edit

  1. ^ Monga I, Kaur K, Dhanda S (March 2022). "Revisiting hematopoiesis: applications of the bulk and single-cell transcriptomics dissecting transcriptional heterogeneity in hematopoietic stem cells". Briefings in Functional Genomics. 21 (3): 159–176. doi:10.1093/bfgp/elac002. PMID 35265979.
  2. ^ a b Birbrair A, Frenette PS (April 2016). "Niche heterogeneity in the bone marrow". Annals of the New York Academy of Sciences. 1370 (1): 82–96. Bibcode:2016NYASA1370...82B. doi:10.1111/nyas.13016. PMC 4938003. PMID 27015419.
  3. ^ a b Dzierzak E, Bigas A (May 2018). "Blood Development: Hematopoietic Stem Cell Dependence and Independence". Cell Stem Cell. 22 (5): 639–651. doi:10.1016/j.stem.2018.04.015. hdl:10230/36965. PMID 29727679.
  4. ^ Ottersbach K (April 2019). "Endothelial-to-haematopoietic transition: an update on the process of making blood". Biochemical Society Transactions. 47 (2): 591–601. doi:10.1042/BST20180320. PMC 6490701. PMID 30902922.
  5. ^ Till JE, McCULLOCH EA (February 1961). "A direct measurement of the radiation sensitivity of normal mouse bone marrow cells". Radiation Research. 14 (2): 213–22. Bibcode:1961RadR...14..213T. doi:10.2307/3570892. hdl:1807/2781. JSTOR 3570892. PMID 13776896.
  6. ^ . Stem Cell Information. National Institutes of Health, U.S. Department of Health and Human Services. 17 June 2011. Archived from the original on 2015-09-29.
  7. ^ Müller, Albrecht M.; Huppertz, Sascha; Henschler, Reinhard (July 2016). "Hematopoietic Stem Cells in Regenerative Medicine: Astray or on the Path?". Transfusion Medicine and Hemotherapy. 43 (4): 247–254. doi:10.1159/000447748. ISSN 1660-3796. PMC 5040947. PMID 27721700.
  8. ^ de Bruijn MF, Speck NA, Peeters MC, Dzierzak E (June 2000). "Definitive hematopoietic stem cells first develop within the major arterial regions of the mouse embryo". The EMBO Journal. 19 (11): 2465–2474. doi:10.1093/emboj/19.11.2465. PMC 212758. PMID 10835345.
  9. ^ Medvinsky A, Dzierzak E (September 1996). "Definitive hematopoiesis is autonomously initiated by the AGM region". Cell. 86 (6): 897–906. doi:10.1016/s0092-8674(00)80165-8. hdl:1765/57137. PMID 8808625. S2CID 3330712.
  10. ^ Ivanovs A, Rybtsov S, Welch L, Anderson RA, Turner ML, Medvinsky A (November 2011). "Highly potent human hematopoietic stem cells first emerge in the intraembryonic aorta-gonad-mesonephros region". The Journal of Experimental Medicine. 208 (12): 2417–2427. doi:10.1084/jem.20111688. PMC 3256972. PMID 22042975.
  11. ^ Dzierzak E, Speck NA (February 2008). "Of lineage and legacy: the development of mammalian hematopoietic stem cells". Nature Immunology. 9 (2): 129–136. doi:10.1038/ni1560. PMC 2696344. PMID 18204427.
  12. ^ "Bone Marrow Transplant Process". Mayo Clinic. Retrieved 18 March 2015.
  13. ^ Mahla RS (2016). "Stem Cells Applications in Regenerative Medicine and Disease Therapeutics". International Journal of Cell Biology. 2016 (7): 6940283. doi:10.1155/2016/6940283. PMC 4969512. PMID 27516776.
  14. ^ Srikanth L, Sunitha MM, Venkatesh K, Kumar PS, Chandrasekhar C, Vengamma B, Sarma PV (2015). "Anaerobic Glycolysis and HIF1α Expression in Haematopoietic Stem Cells Explains Its Quiescence Nature". Journal of Stem Cells. 10 (2): 97–106. PMID 27125138.
  15. ^ a b Baumgartner C, Toifl S, Farlik M, Halbritter F, Scheicher R, Fischer I, et al. (June 2018). "An ERK-Dependent Feedback Mechanism Prevents Hematopoietic Stem Cell Exhaustion". Cell Stem Cell. 22 (6): 879–892.e6. doi:10.1016/j.stem.2018.05.003. PMC 5988582. PMID 29804890.
  16. ^ a b Felfly H, Haddad GG (2014). "Hematopoietic stem cells: potential new applications for translational medicine". Journal of Stem Cells. 9 (3): 163–197. PMID 25157450.
  17. ^ a b c d Park B, Yoo KH, Kim C (December 2015). "Hematopoietic stem cell expansion and generation: the ways to make a breakthrough". Blood Research. 50 (4): 194–203. doi:10.5045/br.2015.50.4.194. PMC 4705045. PMID 26770947.
  18. ^ Mahla RS (2016). "Stem Cells Applications in Regenerative Medicine and Disease Therapeutics". International Journal of Cell Biology. 2016 (7): 6940283. doi:10.1155/2016/6940283. PMC 4969512. PMID 27516776.
  19. ^ . stemcells.nih.gov. Archived from the original on 2021-05-01. Retrieved 2018-01-18.
  20. ^ Dzierzak E, Speck NA (February 2008). "Of lineage and legacy: the development of mammalian hematopoietic stem cells". Nature Immunology. 9 (2): 129–136. doi:10.1038/ni1560. PMC 2696344. PMID 18204427.
  21. ^ Tyndall A, Fassas A, Passweg J, Ruiz de Elvira C, Attal M, Brooks P, et al. (October 1999). "Autologous haematopoietic stem cell transplants for autoimmune disease--feasibility and transplant-related mortality. Autoimmune Disease and Lymphoma Working Parties of the European Group for Blood and Marrow Transplantation, the European League Against Rheumatism and the International Stem Cell Project for Autoimmune Disease". Bone Marrow Transplantation. 24 (7): 729–734. doi:10.1038/sj.bmt.1701987. PMID 10516675.
  22. ^ Burt RK, Loh Y, Pearce W, Beohar N, Barr WG, Craig R, et al. (February 2008). "Clinical applications of blood-derived and marrow-derived stem cells for nonmalignant diseases". JAMA. 299 (8): 925–936. doi:10.1001/jama.299.8.925. PMID 18314435.
  23. ^ El-Sobky TA, El-Haddad A, Elsobky E, Elsayed SM, Sakr HM (March 2017). "Reversal of skeletal radiographic pathology in a case of malignant infantile osteopetrosis following hematopoietic stem cell transplantation". The Egyptian Journal of Radiology and Nuclear Medicine. 48 (1): 237–243. doi:10.1016/j.ejrnm.2016.12.013.
  24. ^ Hashemi Taheri AP, Radmard AR, Kooraki S, Behfar M, Pak N, Hamidieh AA, Ghavamzadeh A (September 2015). "Radiologic resolution of malignant infantile osteopetrosis skeletal changes following hematopoietic stem cell transplantation". Pediatric Blood & Cancer. 62 (9): 1645–1649. doi:10.1002/pbc.25524. PMID 25820806. S2CID 11287381.
  25. ^ Langereis EJ, den Os MM, Breen C, Jones SA, Knaven OC, Mercer J, et al. (March 2016). "Progression of Hip Dysplasia in Mucopolysaccharidosis Type I Hurler After Successful Hematopoietic Stem Cell Transplantation". The Journal of Bone and Joint Surgery. American Volume. 98 (5): 386–395. doi:10.2106/JBJS.O.00601. PMID 26935461.
  26. ^ Müller, Albrecht M.; Huppertz, Sascha; Henschler, Reinhard (2016-07-26). "Hematopoietic Stem Cells in Regenerative Medicine: Astray or on the Path?". Transfusion Medicine and Hemotherapy. 43 (4): 247–254. doi:10.1159/000447748. ISSN 1660-3796. PMC 5040947.
  27. ^ a b Beerman I, Seita J, Inlay MA, Weissman IL, Rossi DJ (July 2014). "Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle". Cell Stem Cell. 15 (1): 37–50. doi:10.1016/j.stem.2014.04.016. PMC 4082747. PMID 24813857.
  28. ^ a b Nijnik A, Woodbine L, Marchetti C, Dawson S, Lambe T, Liu C, et al. (June 2007). "DNA repair is limiting for haematopoietic stem cells during ageing". Nature. 447 (7145): 686–690. Bibcode:2007Natur.447..686N. doi:10.1038/nature05875. PMID 17554302. S2CID 4332976.
  29. ^ Tilgner K, Neganova I, Moreno-Gimeno I, Al-Aama JY, Burks D, Yung S, et al. (August 2013). "A human iPSC model of Ligase IV deficiency reveals an important role for NHEJ-mediated-DSB repair in the survival and genomic stability of induced pluripotent stem cells and emerging haematopoietic progenitors". Cell Death and Differentiation. 20 (8): 1089–1100. doi:10.1038/cdd.2013.44. PMC 3705601. PMID 23722522.
  30. ^ Lucas D, Escudero B, Ligos JM, Segovia JC, Estrada JC, Terrados G, et al. (February 2009). "Altered hematopoiesis in mice lacking DNA polymerase mu is due to inefficient double-strand break repair". PLOS Genetics. 5 (2): e1000389. doi:10.1371/journal.pgen.1000389. PMC 2638008. PMID 19229323.
  31. ^ Avagyan S, Churchill M, Yamamoto K, Crowe JL, Li C, Lee BJ, et al. (September 2014). "Hematopoietic stem cell dysfunction underlies the progressive lymphocytopenia in XLF/Cernunnos deficiency". Blood. 124 (10): 1622–1625. doi:10.1182/blood-2014-05-574863. PMC 4155271. PMID 25075129.
  32. ^ a b Tilgner K, Neganova I, Singhapol C, Saretzki G, Al-Aama JY, Evans J, et al. (September 2013). "Brief report: a human induced pluripotent stem cell model of cernunnos deficiency reveals an important role for XLF in the survival of the primitive hematopoietic progenitors". Stem Cells. 31 (9): 2015–2023. doi:10.1002/stem.1456. PMID 23818183. S2CID 3623309.
  33. ^ Rossi DJ, Bryder D, Seita J, Nussenzweig A, Hoeijmakers J, Weissman IL (June 2007). "Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age". Nature. 447 (7145): 725–729. Bibcode:2007Natur.447..725R. doi:10.1038/nature05862. PMID 17554309. S2CID 4416445.
  34. ^ "Research may reveal why people can suddenly become frail in their 70s". The Guardian. 1 June 2022. Retrieved 18 July 2022.
  35. ^ Mitchell, Emily; Spencer Chapman, Michael; Williams, Nicholas; Dawson, Kevin J.; Mende, Nicole; Calderbank, Emily F.; Jung, Hyunchul; Mitchell, Thomas; Coorens, Tim H. H.; Spencer, David H.; Machado, Heather; Lee-Six, Henry; Davies, Megan; Hayler, Daniel; Fabre, Margarete A.; Mahbubani, Krishnaa; Abascal, Federico; Cagan, Alex; Vassiliou, George S.; Baxter, Joanna; Martincorena, Inigo; Stratton, Michael R.; Kent, David G.; Chatterjee, Krishna; Parsy, Kourosh Saeb; Green, Anthony R.; Nangalia, Jyoti; Laurenti, Elisa; Campbell, Peter J. (June 2022). "Clonal dynamics of haematopoiesis across the human lifespan". Nature. 606 (7913): 343–350. Bibcode:2022Natur.606..343M. doi:10.1038/s41586-022-04786-y. ISSN 1476-4687. PMC 9177428. PMID 35650442.
  36. ^ Cho, Rebecca H; Sieburg, Hans B; Muller-Sieburg, Christa E (2008). "A new mechanism for the aging of hematopoietic stem cells: aging changes the clonal composition of the stem cell compartment but not individual stem cells". Blood. 111 (12): 5553–5561. doi:10.1182/blood-2007-11-123547. PMC 2424153. PMID 18413859.
  37. ^ Llames, Sara; Garcia-Perez, Eva; Meana, Alvaro; Larcher, Fernando; del Rio, Marcela (2015). "Feeder Layer Cell Actions and Applications". Tissue Eng Part B Rev. 21 (4): 345–353. doi:10.1089/ten.teb.2014.0547. PMC 4533020. PMID 25659081.
  38. ^ Burger JA, Spoo A, Dwenger A, Burger M, Behringer D (August 2003). "CXCR4 chemokine receptors (CD184) and alpha4beta1 integrins mediate spontaneous migration of human CD34+ progenitors and acute myeloid leukaemia cells beneath marrow stromal cells (pseudoemperipolesis)". British Journal of Haematology. 122 (4): 579–89. doi:10.1046/j.1365-2141.2003.04466.x. PMID 12899713. S2CID 8764752.
  39. ^ Sieburg HB, Müller-Sieburg CE (2004). "Classification of short kinetics by shape". In Silico Biology. 4 (2): 209–17. PMID 15107024.
  40. ^ Challen, Grant A.; Boles, Nathan; Lin, Kuan-Yin K.; Goodell, Margaret A. (January 2009). "Mouse hematopoietic stem cell identification and analysis". Cytometry Part A. 75A (1): 14–24. doi:10.1002/cyto.a.20674. PMC 2640229. PMID 19023891.
  41. ^ Müller-Sieburg CE, Cho RH, Thoman M, Adkins B, Sieburg HB (August 2002). "Deterministic regulation of hematopoietic stem cell self-renewal and differentiation". Blood. 100 (4): 1302–9. doi:10.1182/blood.V100.4.1302.h81602001302_1302_1309. PMID 12149211.
  42. ^ a b Muller-Sieburg CE, Cho RH, Karlsson L, Huang JF, Sieburg HB (June 2004). "Myeloid-biased hematopoietic stem cells have extensive self-renewal capacity but generate diminished lymphoid progeny with impaired IL-7 responsiveness". Blood. 103 (11): 4111–8. doi:10.1182/blood-2003-10-3448. PMID 14976059.
  43. ^ Sieburg HB, Cho RH, Dykstra B, Uchida N, Eaves CJ, Muller-Sieburg CE (March 2006). "The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets". Blood. 107 (6): 2311–6. doi:10.1182/blood-2005-07-2970. PMC 1456063. PMID 16291588.
  44. ^ Schroeder T (March 2010). "Hematopoietic stem cell heterogeneity: subtypes, not unpredictable behavior". Cell Stem Cell. 6 (3): 203–7. doi:10.1016/j.stem.2010.02.006. PMID 20207223.
  45. ^ Dykstra B, Kent D, Bowie M, McCaffrey L, Hamilton M, Lyons K, et al. (August 2007). "Long-term propagation of distinct hematopoietic differentiation programs in vivo". Cell Stem Cell. 1 (2): 218–29. doi:10.1016/j.stem.2007.05.015. PMID 18371352.
  46. ^ Challen GA, Boles NC, Chambers SM, Goodell MA (March 2010). "Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1". Cell Stem Cell. 6 (3): 265–78. doi:10.1016/j.stem.2010.02.002. PMC 2837284. PMID 20207229.
  47. ^ Flexner SB, Hauck LC, eds. (1993). Random House Unabridged Dictionary (2nd ed.). New York City: Random House. p. 890. ISBN 0-679-42917-4.

External links edit

  • Fact sheet about blood stem cells on EuroStemCell

hematopoietic, stem, cell, this, article, technical, most, readers, understand, please, help, improve, make, understandable, experts, without, removing, technical, details, august, 2015, learn, when, remove, this, message, hscs, stem, cells, that, give, rise, . This article may be too technical for most readers to understand Please help improve it to make it understandable to non experts without removing the technical details August 2015 Learn how and when to remove this message Hematopoietic stem cells HSCs are the stem cells 1 that give rise to other blood cells This process is called haematopoiesis 2 In vertebrates the first definitive HSCs arise from the ventral endothelial wall of the embryonic aorta within the midgestational aorta gonad mesonephros region through a process known as endothelial to hematopoietic transition 3 4 In adults haematopoiesis occurs in the red bone marrow in the core of most bones The red bone marrow is derived from the layer of the embryo called the mesoderm Hematopoietic stem cellOverview of normal human haematopoiesisDetailsPrecursorHemangioblastSystemHematopoietic systemLocationBone marrowFunctionStem cells that give rise to other blood cellsIdentifiersLatincellula haematopoietica praecursoriaAcronym s HSCMeSHD006412THH2 00 01 0 00006Anatomical terms of microanatomy edit on Wikidata Haematopoiesis is the process by which all mature blood cells are produced It must balance enormous production needs the average person produces more than 500 billion blood cells every day with the need to regulate the number of each blood cell type in the circulation In vertebrates the vast majority of hematopoiesis occurs in the bone marrow and is derived from a limited number of hematopoietic stem cells that are multipotent and capable of extensive self renewal Hematopoietic stem cells give rise to different types of blood cells in lines called myeloid and lymphoid Myeloid and lymphoid lineages both are involved in dendritic cell formation Myeloid cells include monocytes macrophages neutrophils basophils eosinophils erythrocytes and megakaryocytes to platelets Lymphoid cells include T cells B cells natural killer cells and innate lymphoid cells The definition of hematopoietic stem cell has developed since they were first discovered in 1961 5 The hematopoietic tissue contains cells with long term and short term regeneration capacities and committed multipotent oligopotent and unipotent progenitors Hematopoietic stem cells constitute 1 10 000 of cells in myeloid tissue HSC transplants are used in the treatment of cancers and other immune system disorders 6 due to their regenerative properties 7 Contents 1 Structure 1 1 Location 1 2 Subtypes 1 3 Isolating stem cells 2 Function 2 1 Haematopoiesis 2 2 Quiescence 2 3 Mobility 3 Clinical significance 3 1 Transplant 3 2 Aging of hematopoietic stem cells 3 2 1 DNA damage 3 2 2 Loss of clonal diversity 4 Research 4 1 Behavior in culture 4 2 Repopulation kinetics 5 Etymology 6 See also 7 References 8 External linksStructure editThey are round non adherent with a rounded nucleus and low cytoplasm to nucleus ratio In shape hematopoietic stem cells resemble lymphocytes Location edit The very first hematopoietic stem cells during mouse and human embryonic development are found in aorta gonad mesonephros region and the vitelline and umbilical arteries 8 9 10 Slightly later HSCs are also found in the placenta yolk sac embryonic head and fetal liver 3 11 Stem and progenitor cells can be taken from the pelvis at the iliac crest using a needle and syringe 12 The cells can be removed as liquid to perform a smear to look at the cell morphology or they can be removed via a core biopsy to maintain the architecture or relationship of the cells to each other and to the bone citation needed Subtypes edit A colony forming unit is a subtype of HSC This sense of the term is different from colony forming units of microbes which is a cell counting unit There are various kinds of HSC colony forming units Colony forming unit granulocyte erythrocyte monocyte megakaryocyte CFU GEMM Colony forming unit lymphocyte CFU L Colony forming unit erythrocyte CFU E Colony forming unit granulocyte macrophage CFU GM Colony forming unit megakaryocyte CFU Meg Colony forming unit basophil CFU B Colony forming unit eosinophil CFU Eos The above CFUs are based on the lineage Another CFU the colony forming unit spleen CFU S was the basis of an in vivo clonal colony formation which depends on the ability of infused bone marrow cells to give rise to clones of maturing hematopoietic cells in the spleens of irradiated mice after 8 to 12 days It was used extensively in early studies but is now considered to measure more mature progenitor or transit amplifying cells rather than stem cells citation needed Isolating stem cells edit Further information Techniques to isolate haematopoietic stem cells Since hematopoietic stem cells cannot be isolated as a pure population it is not possible to identify them in a microscope citation needed Hematopoietic stem cells can be identified or isolated by the use of flow cytometry where the combination of several different cell surface markers particularly CD34 are used to separate the rare hematopoietic stem cells from the surrounding blood cells Hematopoietic stem cells lack expression of mature blood cell markers and are thus called Lin Lack of expression of lineage markers is used in combination with detection of several positive cell surface markers to isolate hematopoietic stem cells In addition hematopoietic stem cells are characterised by their small size and low staining with vital dyes such as rhodamine 123 rhodamine lo or Hoechst 33342 side population Function edit nbsp Diagram of cells that arise from Hematopoietic stem cells during the process of hematopoiesis Haematopoiesis edit Main article HaematopoiesisHematopoietic stem cells are essential to haematopoiesis the formation of the cells within blood Hematopoietic stem cells can replenish all blood cell types i e are multipotent and self renew A small number of hematopoietic stem cells can expand to generate a very large number of daughter hematopoietic stem cells This phenomenon is used in bone marrow transplantation 13 when a small number of hematopoietic stem cells reconstitute the hematopoietic system This process indicates that subsequent to bone marrow transplantation symmetrical cell divisions into two daughter hematopoietic stem cells must occur Stem cell self renewal is thought to occur in the stem cell niche in the bone marrow and it is reasonable to assume that key signals present in this niche will be important in self renewal 2 There is much interest in the environmental and molecular requirements for HSC self renewal as understanding the ability of HSC to replenish themselves will eventually allow the generation of expanded populations of HSC in vitro that can be used therapeutically Quiescence edit Hematopoietic stem cells like all adult stem cells mostly exist in a state of quiescence or reversible growth arrest The altered metabolism of quiescent HSCs helps the cells survive for extended periods of time in the hypoxic bone marrow environment 14 When provoked by cell death or damage Hematopoietic stem cells exit quiescence and begin actively dividing again The transition from dormancy to propagation and back is regulated by the MEK ERK pathway and PI3K AKT mTOR pathway 15 Dysregulation of these transitions can lead to stem cell exhaustion or the gradual loss of active Hematopoietic stem cells in the blood system 15 Mobility edit Hematopoietic stem cells have a higher potential than other immature blood cells to pass the bone marrow barrier and thus may travel in the blood from the bone marrow in one bone to another bone If they settle in the thymus they may develop into T cells In the case of fetuses and other extramedullary hematopoiesis Hematopoietic stem cells may also settle in the liver or spleen and develop This enables Hematopoietic stem cells to be harvested directly from the blood Clinical significance editTransplant edit Main article Hematopoietic stem cell transplantation Hematopoietic stem cell transplantation HSCT is the transplantation of multipotent hematopoietic stem cells usually derived from bone marrow peripheral blood or umbilical cord blood 16 17 18 It may be autologous the patient s own stem cells are used allogeneic the stem cells come from a donor or syngeneic from an identical twin 16 17 It is most often performed for patients with certain cancers of the blood or bone marrow such as multiple myeloma or leukemia 17 In these cases the recipient s immune system is usually destroyed with radiation or chemotherapy before the transplantation Infection and graft versus host disease are major complications of allogeneic HSCT 17 In order to harvest stem cells from the circulating peripheral blood blood donors are injected with a cytokine such as granulocyte colony stimulating factor G CSF that induces cells to leave the bone marrow and circulate in the blood vessels 19 In mammalian embryology the first definitive Hematopoietic stem cells are detected in the AGM aorta gonad mesonephros and then massively expanded in the fetal liver prior to colonising the bone marrow before birth 20 Hematopoietic stem cell transplantation remains a dangerous procedure with many possible complications it is reserved for patients with life threatening diseases As survival following the procedure has increased its use has expanded beyond cancer to autoimmune diseases 21 22 and hereditary skeletal dysplasias notably malignant infantile osteopetrosis 23 24 and mucopolysaccharidosis 25 Stem cells can be used to regenerate different types of tissues HCT is an established as therapy for chronic myeloid leukemia acute lymphatic leukemia aplastic anemia and hemoglobinopathies in addition to acute myeloid leukemia and primary immune deficiencies Hematopoietic system regeneration is typically achieved within 2 4 weeks post chemo or irradiation therapy and HCT HSCs are being clinically tested for their use in non hematopoietic tissue regeneration 26 Aging of hematopoietic stem cells edit DNA damage edit DNA strand breaks accumulate in long term hematopoietic stem cells during aging 27 This accumulation is associated with a broad attenuation of DNA repair and response pathways that depends on HSC quiescence 27 Non homologous end joining NHEJ is a pathway that repairs double strand breaks in DNA NHEJ is referred to as non homologous because the break ends are directly ligated without the need for a homologous template The NHEJ pathway depends on several proteins including ligase 4 DNA polymerase mu and NHEJ factor 1 NHEJ1 also known as Cernunnos or XLF DNA ligase 4 Lig4 has a highly specific role in the repair of double strand breaks by NHEJ Lig4 deficiency in the mouse causes a progressive loss of hematopoietic stem cells during aging 28 Deficiency of lig4 in pluripotent stem cells results in accumulation of DNA double strand breaks and enhanced apoptosis 29 In polymerase mu mutant mice hematopoietic cell development is defective in several peripheral and bone marrow cell populations with about a 40 decrease in bone marrow cell number that includes several hematopoietic lineages 30 Expansion potential of hematopoietic progenitor cells is also reduced These characteristics correlate with reduced ability to repair double strand breaks in hematopoietic tissue Deficiency of NHEJ factor 1 in mice leads to premature aging of hematopoietic stem cells as indicated by several lines of evidence including evidence that long term repopulation is defective and worsens over time 31 Using a human induced pluripotent stem cell model of NHEJ1 deficiency it was shown that NHEJ1 has an important role in promoting survival of the primitive hematopoietic progenitors 32 These NHEJ1 deficient cells possess a weak NHEJ1 mediated repair capacity that is apparently incapable of coping with DNA damages induced by physiological stress normal metabolism and ionizing radiation 32 The sensitivity of hematopoietic stem cells to Lig4 DNA polymerase mu and NHEJ1 deficiency suggests that NHEJ is a key determinant of the ability of stem cells to maintain themselves against physiological stress over time 28 Rossi et al 33 found that endogenous DNA damage accumulates with age even in wild type Hematopoietic stem cells and suggested that DNA damage accrual may be an important physiological mechanism of stem cell aging Loss of clonal diversity edit A study shows the clonal diversity of hematopoietic stem cells gets drastically reduced around age 70 to a faster growing few substantiating a novel theory of ageing which could enable healthy aging 34 35 Of note the shift in clonal diversity during aging was previously reported in 2008 36 for the murine system by the Christa Muller Sieburg laboratory in San Diego California Research editBehavior in culture edit A cobblestone area forming cell CAFC assay is a cell culture based empirical assay When plated onto a confluent culture of stromal feeder layer 37 a fraction of hematopoietic stem cells creep between the gaps even though the stromal cells are touching each other and eventually settle between the stromal cells and the substratum here the dish surface or trapped in the cellular processes between the stromal cells Emperipolesis is the in vivo phenomenon in which one cell is completely engulfed into another e g thymocytes into thymic nurse cells on the other hand when in vitro lymphoid lineage cells creep beneath nurse like cells the process is called pseudoemperipolesis This similar phenomenon is more commonly known in the HSC field by the cell culture terminology cobble stone area forming cells CAFC which means areas or clusters of cells look dull cobblestone like under phase contrast microscopy compared to the other hematopoietic stem cells which are refractile This happens because the cells that are floating loosely on top of the stromal cells are spherical and thus refractile However the cells that creep beneath the stromal cells are flattened and thus not refractile The mechanism of pseudoemperipolesis is only recently coming to light It may be mediated by interaction through CXCR4 CD184 the receptor for CXC Chemokines e g SDF1 and a4b1 integrins 38 Repopulation kinetics edit Hematopoietic stem cells HSC cannot be easily observed directly and therefore their behaviors need to be inferred indirectly Clonal studies are likely the closest technique for single cell in vivo studies of HSC Here sophisticated experimental and statistical methods are used to ascertain that with a high probability a single HSC is contained in a transplant administered to a lethally irradiated host The clonal expansion of this stem cell can then be observed over time by monitoring the percent donor type cells in blood as the host is reconstituted The resulting time series is defined as the repopulation kinetic of the HSC The reconstitution kinetics are very heterogeneous However using symbolic dynamics one can show that they fall into a limited number of classes 39 To prove this several hundred experimental repopulation kinetics from clonal Thy 1lo SCA 1 lin B220 CD4 CD8 Gr 1 Mac 1 and Ter 119 40 c kit HSC were translated into symbolic sequences by assigning the symbols whenever two successive measurements of the percent donor type cells have a positive negative or unchanged slope respectively By using the Hamming distance the repopulation patterns were subjected to cluster analysis yielding 16 distinct groups of kinetics To finish the empirical proof the Laplace add one approach was used to determine that the probability of finding kinetics not contained in these 16 groups is very small By corollary this result shows that the hematopoietic stem cell compartment is also heterogeneous by dynamical criteria It was originally believed that all hematopoietic stem cells were alike in their self renewal and differentiation abilities This view was first challenged by the 2002 discovery by the Muller Sieburg group in San Diego who illustrated that different stem cells can show distinct repopulation patterns that are epigenetically predetermined intrinsic properties of clonal Thy 1lo Sca 1 lin c kit HSC 41 42 43 The results of these clonal studies led to the notion of lineage bias Using the ratio r L M displaystyle rho L M nbsp of lymphoid L to myeloid M cells in blood as a quantitative marker the stem cell compartment can be split into three categories of HSC Balanced Bala hematopoietic stem cells repopulate peripheral white blood cells in the same ratio of myeloid to lymphoid cells as seen in unmanipulated mice on average about 15 myeloid and 85 lymphoid cells or 3 r 10 Myeloid biased My bi hematopoietic stem cells give rise to very few lymphocytes resulting in ratios 0 lt r lt 3 while lymphoid biased Ly bi hematopoietic stem cellsgenerate very few myeloid cells which results in lymphoid to myeloid ratios of r gt 10 All three types are normal types of HSC and they do not represent stages of differentiation Rather these are three classes of HSC each with an epigenetically fixed differentiation program These studies also showed that lineage bias is not stochastically regulated or dependent on differences in environmental influence My bi HSC self renew longer than balanced or Ly bi HSC The myeloid bias results from reduced responsiveness to the lymphopoetin interleukin 7 IL 7 42 Subsequently other groups confirmed and highlighted the original findings 44 For example the Eaves group confirmed in 2007 that repopulation kinetics long term self renewal capacity and My bi and Ly bi are stably inherited intrinsic HSC properties 45 In 2010 the Goodell group provided additional insights about the molecular basis of lineage bias in side population SP SCA 1 lin c kit HSC 46 As previously shown for IL 7 signaling it was found that a member of the transforming growth factor family TGF beta induces and inhibits the proliferation of My bi and Ly bi HSC respectively Etymology editFrom Greek haimato combining form of haima blood and from the Latinized form of Greek poietikos capable of making creative productive from poiein to make create 47 See also editHematopoietic stem cell niche List of distinct cell types in the adult human bodyReferences edit Monga I Kaur K Dhanda S March 2022 Revisiting hematopoiesis applications of the bulk and single cell transcriptomics dissecting transcriptional heterogeneity in hematopoietic stem cells Briefings in Functional Genomics 21 3 159 176 doi 10 1093 bfgp elac002 PMID 35265979 a b Birbrair A Frenette PS April 2016 Niche heterogeneity in the bone marrow Annals of the New York Academy of Sciences 1370 1 82 96 Bibcode 2016NYASA1370 82B doi 10 1111 nyas 13016 PMC 4938003 PMID 27015419 a b Dzierzak E Bigas A May 2018 Blood Development Hematopoietic Stem Cell Dependence and Independence Cell Stem Cell 22 5 639 651 doi 10 1016 j stem 2018 04 015 hdl 10230 36965 PMID 29727679 Ottersbach K April 2019 Endothelial to haematopoietic transition an update on the process of making blood Biochemical Society Transactions 47 2 591 601 doi 10 1042 BST20180320 PMC 6490701 PMID 30902922 Till JE McCULLOCH EA February 1961 A direct measurement of the radiation sensitivity of normal mouse bone marrow cells Radiation Research 14 2 213 22 Bibcode 1961RadR 14 213T doi 10 2307 3570892 hdl 1807 2781 JSTOR 3570892 PMID 13776896 5 Hematopoietic Stem Cells Stem Cell Information National Institutes of Health U S Department of Health and Human Services 17 June 2011 Archived from the original on 2015 09 29 Muller Albrecht M Huppertz Sascha Henschler Reinhard July 2016 Hematopoietic Stem Cells in Regenerative Medicine Astray or on the Path Transfusion Medicine and Hemotherapy 43 4 247 254 doi 10 1159 000447748 ISSN 1660 3796 PMC 5040947 PMID 27721700 de Bruijn MF Speck NA Peeters MC Dzierzak E June 2000 Definitive hematopoietic stem cells first develop within the major arterial regions of the mouse embryo The EMBO Journal 19 11 2465 2474 doi 10 1093 emboj 19 11 2465 PMC 212758 PMID 10835345 Medvinsky A Dzierzak E September 1996 Definitive hematopoiesis is autonomously initiated by the AGM region Cell 86 6 897 906 doi 10 1016 s0092 8674 00 80165 8 hdl 1765 57137 PMID 8808625 S2CID 3330712 Ivanovs A Rybtsov S Welch L Anderson RA Turner ML Medvinsky A November 2011 Highly potent human hematopoietic stem cells first emerge in the intraembryonic aorta gonad mesonephros region The Journal of Experimental Medicine 208 12 2417 2427 doi 10 1084 jem 20111688 PMC 3256972 PMID 22042975 Dzierzak E Speck NA February 2008 Of lineage and legacy the development of mammalian hematopoietic stem cells Nature Immunology 9 2 129 136 doi 10 1038 ni1560 PMC 2696344 PMID 18204427 Bone Marrow Transplant Process Mayo Clinic Retrieved 18 March 2015 Mahla RS 2016 Stem Cells Applications in Regenerative Medicine and Disease Therapeutics International Journal of Cell Biology 2016 7 6940283 doi 10 1155 2016 6940283 PMC 4969512 PMID 27516776 Srikanth L Sunitha MM Venkatesh K Kumar PS Chandrasekhar C Vengamma B Sarma PV 2015 Anaerobic Glycolysis and HIF1a Expression in Haematopoietic Stem Cells Explains Its Quiescence Nature Journal of Stem Cells 10 2 97 106 PMID 27125138 a b Baumgartner C Toifl S Farlik M Halbritter F Scheicher R Fischer I et al June 2018 An ERK Dependent Feedback Mechanism Prevents Hematopoietic Stem Cell Exhaustion Cell Stem Cell 22 6 879 892 e6 doi 10 1016 j stem 2018 05 003 PMC 5988582 PMID 29804890 a b Felfly H Haddad GG 2014 Hematopoietic stem cells potential new applications for translational medicine Journal of Stem Cells 9 3 163 197 PMID 25157450 a b c d Park B Yoo KH Kim C December 2015 Hematopoietic stem cell expansion and generation the ways to make a breakthrough Blood Research 50 4 194 203 doi 10 5045 br 2015 50 4 194 PMC 4705045 PMID 26770947 Mahla RS 2016 Stem Cells Applications in Regenerative Medicine and Disease Therapeutics International Journal of Cell Biology 2016 7 6940283 doi 10 1155 2016 6940283 PMC 4969512 PMID 27516776 5 Hematopoietic Stem Cells stemcells nih gov stemcells nih gov Archived from the original on 2021 05 01 Retrieved 2018 01 18 Dzierzak E Speck NA February 2008 Of lineage and legacy the development of mammalian hematopoietic stem cells Nature Immunology 9 2 129 136 doi 10 1038 ni1560 PMC 2696344 PMID 18204427 Tyndall A Fassas A Passweg J Ruiz de Elvira C Attal M Brooks P et al October 1999 Autologous haematopoietic stem cell transplants for autoimmune disease feasibility and transplant related mortality Autoimmune Disease and Lymphoma Working Parties of the European Group for Blood and Marrow Transplantation the European League Against Rheumatism and the International Stem Cell Project for Autoimmune Disease Bone Marrow Transplantation 24 7 729 734 doi 10 1038 sj bmt 1701987 PMID 10516675 Burt RK Loh Y Pearce W Beohar N Barr WG Craig R et al February 2008 Clinical applications of blood derived and marrow derived stem cells for nonmalignant diseases JAMA 299 8 925 936 doi 10 1001 jama 299 8 925 PMID 18314435 El Sobky TA El Haddad A Elsobky E Elsayed SM Sakr HM March 2017 Reversal of skeletal radiographic pathology in a case of malignant infantile osteopetrosis following hematopoietic stem cell transplantation The Egyptian Journal of Radiology and Nuclear Medicine 48 1 237 243 doi 10 1016 j ejrnm 2016 12 013 Hashemi Taheri AP Radmard AR Kooraki S Behfar M Pak N Hamidieh AA Ghavamzadeh A September 2015 Radiologic resolution of malignant infantile osteopetrosis skeletal changes following hematopoietic stem cell transplantation Pediatric Blood amp Cancer 62 9 1645 1649 doi 10 1002 pbc 25524 PMID 25820806 S2CID 11287381 Langereis EJ den Os MM Breen C Jones SA Knaven OC Mercer J et al March 2016 Progression of Hip Dysplasia in Mucopolysaccharidosis Type I Hurler After Successful Hematopoietic Stem Cell Transplantation The Journal of Bone and Joint Surgery American Volume 98 5 386 395 doi 10 2106 JBJS O 00601 PMID 26935461 Muller Albrecht M Huppertz Sascha Henschler Reinhard 2016 07 26 Hematopoietic Stem Cells in Regenerative Medicine Astray or on the Path Transfusion Medicine and Hemotherapy 43 4 247 254 doi 10 1159 000447748 ISSN 1660 3796 PMC 5040947 a b Beerman I Seita J Inlay MA Weissman IL Rossi DJ July 2014 Quiescent hematopoietic stem cells accumulate DNA damage during aging that is repaired upon entry into cell cycle Cell Stem Cell 15 1 37 50 doi 10 1016 j stem 2014 04 016 PMC 4082747 PMID 24813857 a b Nijnik A Woodbine L Marchetti C Dawson S Lambe T Liu C et al June 2007 DNA repair is limiting for haematopoietic stem cells during ageing Nature 447 7145 686 690 Bibcode 2007Natur 447 686N doi 10 1038 nature05875 PMID 17554302 S2CID 4332976 Tilgner K Neganova I Moreno Gimeno I Al Aama JY Burks D Yung S et al August 2013 A human iPSC model of Ligase IV deficiency reveals an important role for NHEJ mediated DSB repair in the survival and genomic stability of induced pluripotent stem cells and emerging haematopoietic progenitors Cell Death and Differentiation 20 8 1089 1100 doi 10 1038 cdd 2013 44 PMC 3705601 PMID 23722522 Lucas D Escudero B Ligos JM Segovia JC Estrada JC Terrados G et al February 2009 Altered hematopoiesis in mice lacking DNA polymerase mu is due to inefficient double strand break repair PLOS Genetics 5 2 e1000389 doi 10 1371 journal pgen 1000389 PMC 2638008 PMID 19229323 Avagyan S Churchill M Yamamoto K Crowe JL Li C Lee BJ et al September 2014 Hematopoietic stem cell dysfunction underlies the progressive lymphocytopenia in XLF Cernunnos deficiency Blood 124 10 1622 1625 doi 10 1182 blood 2014 05 574863 PMC 4155271 PMID 25075129 a b Tilgner K Neganova I Singhapol C Saretzki G Al Aama JY Evans J et al September 2013 Brief report a human induced pluripotent stem cell model of cernunnos deficiency reveals an important role for XLF in the survival of the primitive hematopoietic progenitors Stem Cells 31 9 2015 2023 doi 10 1002 stem 1456 PMID 23818183 S2CID 3623309 Rossi DJ Bryder D Seita J Nussenzweig A Hoeijmakers J Weissman IL June 2007 Deficiencies in DNA damage repair limit the function of haematopoietic stem cells with age Nature 447 7145 725 729 Bibcode 2007Natur 447 725R doi 10 1038 nature05862 PMID 17554309 S2CID 4416445 Research may reveal why people can suddenly become frail in their 70s The Guardian 1 June 2022 Retrieved 18 July 2022 Mitchell Emily Spencer Chapman Michael Williams Nicholas Dawson Kevin J Mende Nicole Calderbank Emily F Jung Hyunchul Mitchell Thomas Coorens Tim H H Spencer David H Machado Heather Lee Six Henry Davies Megan Hayler Daniel Fabre Margarete A Mahbubani Krishnaa Abascal Federico Cagan Alex Vassiliou George S Baxter Joanna Martincorena Inigo Stratton Michael R Kent David G Chatterjee Krishna Parsy Kourosh Saeb Green Anthony R Nangalia Jyoti Laurenti Elisa Campbell Peter J June 2022 Clonal dynamics of haematopoiesis across the human lifespan Nature 606 7913 343 350 Bibcode 2022Natur 606 343M doi 10 1038 s41586 022 04786 y ISSN 1476 4687 PMC 9177428 PMID 35650442 Cho Rebecca H Sieburg Hans B Muller Sieburg Christa E 2008 A new mechanism for the aging of hematopoietic stem cells aging changes the clonal composition of the stem cell compartment but not individual stem cells Blood 111 12 5553 5561 doi 10 1182 blood 2007 11 123547 PMC 2424153 PMID 18413859 Llames Sara Garcia Perez Eva Meana Alvaro Larcher Fernando del Rio Marcela 2015 Feeder Layer Cell Actions and Applications Tissue Eng Part B Rev 21 4 345 353 doi 10 1089 ten teb 2014 0547 PMC 4533020 PMID 25659081 Burger JA Spoo A Dwenger A Burger M Behringer D August 2003 CXCR4 chemokine receptors CD184 and alpha4beta1 integrins mediate spontaneous migration of human CD34 progenitors and acute myeloid leukaemia cells beneath marrow stromal cells pseudoemperipolesis British Journal of Haematology 122 4 579 89 doi 10 1046 j 1365 2141 2003 04466 x PMID 12899713 S2CID 8764752 Sieburg HB Muller Sieburg CE 2004 Classification of short kinetics by shape In Silico Biology 4 2 209 17 PMID 15107024 Challen Grant A Boles Nathan Lin Kuan Yin K Goodell Margaret A January 2009 Mouse hematopoietic stem cell identification and analysis Cytometry Part A 75A 1 14 24 doi 10 1002 cyto a 20674 PMC 2640229 PMID 19023891 Muller Sieburg CE Cho RH Thoman M Adkins B Sieburg HB August 2002 Deterministic regulation of hematopoietic stem cell self renewal and differentiation Blood 100 4 1302 9 doi 10 1182 blood V100 4 1302 h81602001302 1302 1309 PMID 12149211 a b Muller Sieburg CE Cho RH Karlsson L Huang JF Sieburg HB June 2004 Myeloid biased hematopoietic stem cells have extensive self renewal capacity but generate diminished lymphoid progeny with impaired IL 7 responsiveness Blood 103 11 4111 8 doi 10 1182 blood 2003 10 3448 PMID 14976059 Sieburg HB Cho RH Dykstra B Uchida N Eaves CJ Muller Sieburg CE March 2006 The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets Blood 107 6 2311 6 doi 10 1182 blood 2005 07 2970 PMC 1456063 PMID 16291588 Schroeder T March 2010 Hematopoietic stem cell heterogeneity subtypes not unpredictable behavior Cell Stem Cell 6 3 203 7 doi 10 1016 j stem 2010 02 006 PMID 20207223 Dykstra B Kent D Bowie M McCaffrey L Hamilton M Lyons K et al August 2007 Long term propagation of distinct hematopoietic differentiation programs in vivo Cell Stem Cell 1 2 218 29 doi 10 1016 j stem 2007 05 015 PMID 18371352 Challen GA Boles NC Chambers SM Goodell MA March 2010 Distinct hematopoietic stem cell subtypes are differentially regulated by TGF beta1 Cell Stem Cell 6 3 265 78 doi 10 1016 j stem 2010 02 002 PMC 2837284 PMID 20207229 Flexner SB Hauck LC eds 1993 Random House Unabridged Dictionary 2nd ed New York City Random House p 890 ISBN 0 679 42917 4 External links editFact sheet about blood stem cells on EuroStemCell Retrieved from https en wikipedia org w index php title Hematopoietic stem cell amp oldid 1222251598, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.