Büşra Nur Barmanbay1, Oytun Erbaş1

1ERBAS Institute of Experimental Medicine, Illinois, USA & Gebze, Türkiye

Keywords: Embryonic stem cells, hematopoietic stem cells, longterm hematopoietic stem cells, mesenchymal stem cells, short-term hematopoietic stem cells.

Abstract

Hematopoietic stem cells (HSCs) are cells that can differentiate into any type of blood cell and can keep on making blood for the rest of a person's life. There is a role of HSCs in blood production and in many cellular processes such as apoptosis, calcium balance, mitochondria, and reactive oxygen species production. Mutations originating in HSCs)underlie genetic blood diseases. Additionally, HSCs are subject to the effects of aging, impairing their ability to maintain cell polarity. In this chapter, we extensively examined the characteristics and implications of HSCs.

Introduction

The term “stem cell (SC)”[1-3] was first used in a scientific publication in 1868 by the German biologist Ernst Haeckel. When referring to the single-celled organism’s ancestor, Haeckel called it a “Stammzelle” (stem cell). In 1892, Valentin Hacker referred to SCs as cells that produce immature egg cells in sexual organs. In the 1960s, Stevens et al.[4] revealed that the embryonal carcinoma cells they studied were actually pluripotent stem cells (PSCs).[5] In the early 1980s, Gail Martins, Martin Evans, and Matthew Kaufman isolated stem cells from mouse embryos and coined the term “embryonic stem cells (ESCs)” in the literature.[6] Jamie Thompson first cultured human ESCs in 1999, following monkey ESCs.[5] Stem cells are valuable for organ formation during growth, helping a living thing’s lifelong tissue function.[7] Stem cells are found in many adult tissues, including blood, umbilical cord blood (UCB), bone marrow (BM), the hematopoietic system, adipose tissue, gametes, the gut, the nervous system, the epidermis, the heart, skeletal muscles.[5,8-10]

Stem cells are one of the main sources of tissue development. In normal processes, SCs are dormant but are activated during certain life cycle processes or in cases of injury.[8] The proliferation and cell cycle differentiation of SCs replaces damaged and aging cells.[11] The ability to reproduce indefinitely is associated with telomerase enzyme activity. The end zone of the deoxyribonucleic acid (DNA) sequence, which regulates the proliferation of cells, has a chain of telomeres. This is slightly different in the SC telomere chain is longer, and telomerase enzyme activity is higher. As a result, SCs can replicate for a long time by self-replication.[12] Basic properties of SCs include self-renewal ability, differentiation (potency) into various cell lineages and tissue types, proliferation capacities, usually arising from a single cell (clonal), unlimited proliferation, and repairing tissue when given to a damaged cell.[5,6,9,13-17]

Stem cells have critical potential in regenerative medicine treatments since they can maintain homeostasis, obtain it easily, regenerate injured or damaged tissue, and can be transferred from the same body to transplanted (autologous) or to different recipients (allogenic) in inheritance within the same species.[5,9,17] Stem cells are called niches, the microenvironment that provides the necessary functions for their growth and change.[9,11] In 1970, the term “SC niche” was proposed for the human hematopoietic system.[11] The idea of being able to regenerate damaged tissue is based on the Titan god of fire Prometheus in ancient Greek mythology. Prometheus was condemned to eternal torment by the Greek god Zeus for stealing fire and giving it to humans. Prometheus is chained to a rock, and Zeus sends an eagle, the symbol of Zeus, to eat Prometheus’ liver every day. Every night the liver regenerates and grows.[16,18] With the ability of SCs to self-replenish and repair damaged tissue offers hope for an entire future recovery from many diseases.[5] Stem cells are divided into five groups according to their differentiation power; totipotent, pluripotent, oligopotent, multipotent, and unipotent.[6,12,14,16,17]

Stem cells are divided into three groups by origin; ESCs, adult stem cells, and induced pluripotent stem cells (iPSCs), as shown in Figure 1.[16]

Totipotent Stem Cells

The fertilized egg cell (zygote) is the first ESC that can change into all the cells in the creation of a living being. These cells are called “totipotent cells” (Totus: not fully divided, potential: power) that can do many things.[12,16,17] Totipotent stem cells, which can form embryonal structures, can change all of the cells required for adulthood.[5,12] Totipotent cells are pluripotent, multipotent, and unipotent stem cells that can give rise to embryonic structures such as the umbilical cord, amniotic sac, umbilical cord gel (Wharton’s gel), and placenta.[6,12,16,17]

Pluripotent Stem Cells

Pluripotent stem cells are found in the blastocyst stage of the embryo.[12] They have the ability to differentiate into all embryonal structures, but they do not form a new living thing.[6,17] They can become an average of 200 cell types under mandatory cells.[12] Pluripotent stem cells can develop into all cell types from the three germ layers, ectoderm, endoderm, and mesoderm, from which all tissues and organs develop.[6,14,16,17] It can be derived from many adult tissues, such as BM.[6]

Multipotent Stem Cells

Multipotent stem cells can differentiate into a single type of germ layer and can be found in many tissues.[5,6,14,16,17] The bottom section of the hierarchical tree has multipotent stem cells.[5] They are cells from other stages of embryonic development.[12] It is isolated from BM, fat tissue, bone, UCB, amniotic sac, gel (Wharton gel) in the umbilical cord, and peripheral blood.[16] Under laboratory conditions, it can differentiate into a plethora of different cell types.[17]

Oligopotent Stem Cells

Oligopotent stem cells have the ability to self-renew. It can form two or more species in a given cell.[6,13,16] The oligopotent stem structure is made up of BM, adipose tissue, and trabecular bone at the ends of the spine’s long bones.[14]

Unipotent Stem Cells

Unipotent stem cells have the ability to self-regenerate and have the ability to change to a limited type of cells.[6,14,16,17] As a result, they are known as stable progenitors since they can support extremely limited cell proliferation (proliferative).[5] It serves in the repair of cells, but PSCs are needed to repair large cell destructions.[17] Unipotent stem cells are derived from BM, fat tissue, and trabecular bone, which is located on the tip of long bones in the vertebrae.[14]

Induced Pluripotent Stem Cells

Induced pluripotent stem cells are derived from reprogrammed matured somatic cells.[14]

They are SCs that grow at speed, they are used in the clinic. Induced pluripotent stem cells are used in areas such as drug development, modeling of diseases, and renewed medicine.[16]

Embryonic Stem Cells

Embryonic stem cells are extracted from a blastocyst, which forms in the uterus 5-6 days after fertilization.[5,14,16,17] Embryonic stem cells have the ability to reproduce infinitely.[17] Embryonic stem cells are defined as pluripotent according to their differentiation power. The ectoderm, where all tissues and organs develop, can be differentiated into three germ layers, the endoderm, and the mesoderm.[5,16,17] Due to legal and ethical rules, the use of ESCs is limited. For this reason, mesenchymal stem cells (MSCs) are used.[14,16]

Adult Stem Cells

Adult stem cells are derived from mature tissues.[16] Adult stem cells can replenish themselves asymmetrically by splitting.[17] They are the most studied multipotent, hematopoietic, and MSCs capable of multipotent from their group in adult stem cells.[17] Although the entire tissue of the three germ layers is isolated, differentiation capacity is limited. Adult stem cells are advantageous from an ethical standpoint. [16]

Mesenchymal Stem Cell

Mesenchymal stem cells[20-22] are derived from the mesoderm layer, neonatal and adult tissues.[14,17] Many types of cells are called MSCs since they can change and multiply under laboratory conditions. Bone marrow, adipose tissue, fatty tissue, and cord blood are all sources of MSC.[17] Today it is used in the treatment of many diseases; It is used in cartilage destruction, calcification (osteoarthritis), bone damage, osteoporosis, and degenerative diseases.[12]

HEMATOPOIETIC STEM CELLS

Hematopoietic stem cells (HSCs) are constantly self-renewing to protect against stem cell accumulation.[23-30] Hematopoietic stem cells are the only SCs that can differentiate into all types of blood cells capable of producing blood for life.[23,24,28,31-34]

Blood is the body fluid that provides the vital functions of a living being.[35,36] The soul was thought to be in the blood of ancient times.[35]

Hematopoiesis, the production of blood cells to keep the fetus alive, occurs in the yolk sac (vitellus sac) on day 27, the third week of the embryo.[29,37-39] The dorsal aorta is the main site where the first HSCs form.[38] Hematopoietic stem cells are stored in the aorta-gonad-mesonephros region, placenta, fetal liver, thymus gland, spleen, and finally BM. [29,40-43]

Hematopoietic stem cells from the aorta-gonad-mesonephros region and the placenta self-renew. Hematopoietic stem cells in the BM and fetal liver can differentiate into all types of blood cells.[29,40,44-46] Mature blood cells are produced by HSCs found in the BM, as shown in Figure 2.[20,25,27,29,35,47,48]

According to the European Blood and Marrow Transplant Group, a single case of death was recorded from the first 27,770 HSC transplants isolated from the BM during the 1993-2005 period.[14] On a daily basis in an adult individual, HSC produces an average of 1011 blood cells.[23] The most commonly studied species of adult tissue-based stem cells are HSCs.[14] The first traces of HSCs II. It emerged as a result of the observation that blood production was negative as a result of exposure to high amounts of radiation as a result of the atomic bombing in World War II.[38] In the late 20th century, HSCs were isolated from BM. They began to be studied on mice with leukemia for use in treatment. With the successful results of these studies, clinical stem cell transplantation started in 1970 the 1980s and is still used in the treatment of blood diseases today.[14,22,25,49-51]

Many genetic blood disorders are caused by point mutations that affect HSCs and their types, resulting in hematopoiesis or cell type errors.[52,53] Hematopoietic stem cells are obtained from BM, peripheral blood, and cord blood.[17,24,25,50,54] They were realized clinical potential early in the history of renewed medicine.[14] In the emerging field of tissue engineering, HSCs are used in BM transplants, gene therapy, and the production of blood products.[55] It’s used as a therapeutic cellular tool for treating blood diseases. The treatment method using hematopoietic stem cells is an HSC transplant (HSCT).[24,36]

There are HSCs in many cellular processes, such as apoptosis, calcium balance, mitochondria, and reactive oxygen species (ROS) production.[34,37,56-58]

In natural conditions and under stress, HSCs are rare (~3,000 to 10,000 per adult human) and are often inactive, moving in motionless, quiet, and active states to meet the needs of the body.[26-30,59-61]

The very low oxygen level (hypoxic) SC niche is the most suitable environment for the long-term preservation and storage of HSCs, as the energy requirement will be low.[56] Bone morphogenetic protein (BMP), Notch, and Wnt signaling pathways are important for the production of HSCs. [36] Cytokines appear to influence HSC survival through transcriptional or translational regulation of molecules involved in apoptosis. It is not known which of the growth factors plays a crucial role in the survival and proliferation of HSCs. In laboratory studies, IL-3, SCF (ligand for c-kit), Flt-3 ligand, and GM-CSF were found to be valuable, but these results imply that cytokines are overworked. Recently, IL-3 has been found to be involved in the survival of HSCs in the region of the mid-gestational anogenital space (AGM).[37,41,62,63]

To date, many surface markers have been identified to obtain more HSCs. These; CD34, membrane glycoprotein Sca-1, tyrosine kinase receptor (CD117) c-Kit, CD150, and signaling lymphocyte activation molecule (SLAM) are still in use in laboratories today.[37,44,49,51,64] According to CD34 tokens, HSC is divided into two subgroups: 1. Long-term HSCs (LT-HSCs), 2.Short-term HSCs (ST-HSCs).[49]

Long-Term Hematopoietic Stem Cells

Long-term hematopoietic stem cells are uncommon and inactive in the BM. [29,39,49] Demonstrates ability to regenerate long term > six months and longer regenerates.[34,49,50] In the G0 cell cycle during blood production, the appropriate extracellular cannot replicate unless they are separated by signals, they have low mitochondria activity.[29,37] The LT-HSCs can be converted to all adult species.[29] It is known that recurrent infections and chronic inflammation reduce the self-renewal ability of LTR-HSCs, resulting in a decrease and extinction of HSC accumulation. [50]

Short-Term Hematopoietic Stem Cells

Short-term hematopoietic stem cells have limited ability to renew themselves.[29,34,50,59] Short Term

AGED HEMATOPOIETIC STEM CELLS

Hematopoietic stem cells are known to age. Studies have shown that HSCs obtained from older mice become weaker than HSCs obtained in young mice.[37] It is known that the density of the aged HSCs[65] HSCs in the BM increases with age and loses their function. Another feature of aged HSCs is the loss of cell polarity. The aging of HSCs results from permanent cell cycle arrest, apoptosis, or accumulative cells that cause senescence, and genomic destruction.[20] The senescence of HSCs indicated by defective signaling pathways, DNA damage, epigenetic differences, and high levels of ROS, as well as senescence-related differences in the structure of the BM niche, were thought to be the main cause of HSC senescence. The aged BM niche influences the functional senescence of young HSCs.[34,66] Since the density of HSCs with low levels of ROS decreases with age, ROS production is the hallmark of aging. Adequate ROS levels are important intermediaries of different signal transmission paths. Increased ROS levels affect the life, self-renewal, and change of HSCs. Reactive oxygen species, causing HSC aging and excessive ROS production stimulate apoptosis in HSCs.[67] The bone marrow is a space for cleaning aged HSCs.[20]

BONE MARROW NICHE

Bone marrow contains a stack of inert multipotent stem cells, the main source for the constant renewal of all blood cell types for life.[66,68] Bone marrow is where SCs that do not belong in the newborn are obtained.[6,14] Hematopoietic stem cells are located in the BM. [51,65,69,70] Inside the BM niche, there are personal HSC niches.[59,64,66] The complex vasculature of the BM consists of different small arteries, which are composed of the feeding artery that directs the blood to the extensive vein network.[66] Stem cell niches in 1970 situated special cells appanage chapters with specific is and at this place motionless by means of halt.[6,59] The niche is a special compartment in the BM that protects HSCs found in the extracellular matrix, blood, and synovial fluid.[6,21,24,30,59,71]

Hematopoietic stem cell niches are located around vessels in the spleen and BM. Endothelial cells and stromal cells secrete factors that support the maintenance and regulation of HSC niches.[72] Important features of HSC niches are cell adhesion, regulation of migration, proliferation, control of differentiation, and determination of cell shape.[71] It has an effect on the formation of many cell HSC niches such as osteoblasts, endothelial cells, CXCL12 abundant reticular (CAR) cells, mesenchymal progenitor cells, myelinated Schwann cells surrounding autonomic nerves, macrophages, megakaryocytes, osteoclasts.[24,72]

In conclusion, HSCs produce blood. In an adult person, HSC can produce an average of 1,011 blood cells. There are HSCs in many cellular processes; apoptosis, calcium balance, mitochondria, and ROS production. The cause of hereditary blood diseases is mutations occurring in HSCs. And the way that these diseases are cured is by transplanting HSC. It is known that HSCs have a role in aging. There are many complex molecular mechanisms in the spatial process and aging of HSCs. Although it’s used today to treat blood disorders, we think it’s possible to use HSC for treating all diseases with advanced technology and molecular mechanisms illuminated every day.

Cite this article as: Barmanbay BN, Erbaş O. Hematopoietic Stem Cells. JEB Med Sci 2024;5(1):12-18.

Conflict of Interest

The authors declared no conflicts of interest with respect to the authorship and/or publication of this article.

Financial Disclosure

The authors received no financial support for the research and/or authorship of this article.

Acknowledgments

The Figures (Figure 1 and Figure 2) used in this chapter were created with BioRender (BioRender.com).

References

  1. Saigal S, Bhargava A. Stem cell--is there any role in the tumorigenic activity. Turk Patoloji Derg. 2011 May;27:93-7.
  2. Akan H. Antifungal prophylaxis in stem cell transplantation centers in Turkey. Turk J Haematol. 2011 Dec 5;28:271-5.
  3. Orford KW, Scadden DT. Deconstructing stem cell selfrenewal: genetic insights into cell-cycle regulation. Nat Rev Genet. 2008 Feb;9:115-28.
  4. Stevens LC. Teratomların biyolojisi. Adv Morphog. 1967;1-31.
  5. K. Appasani, R.K. Appasani. Stem cell biology and regenerative medicine. Springer Science+Business Media. 2011 Oct;3-19.
  6. Lupatov AY, Yarygin KN. Telomeres and Telomerase in the Control of Stem Cells. Biomedicines. 2022 Sep 20;10:2335.
  7. Huang HT, Zon LI. Regulation of stem cells in the zebrafish hematopoietic system. Cold Spring Harb Symp Quant Biol. 2008;73:111-8.
  8. Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that promote stem cell maintenance throughout life. Cell. 2008 Feb 22;132:598-611.
  9. Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell. 2004 Sep 3;118:635-48.
  10. Mimeault M, Batra SK. Concise review: recent advances on the significance of stem cells in tissue regeneration and cancer therapies. Stem Cells. 2006 Nov;24:2319-45.
  11. Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell. 2004 Jul 23;118:149-61.
  12. Barmanbay BN, Altuntaş İ, Erbaş O. The Role of Serotonin in Breast Cancer. JEB Med Sci 2022;3:221-6.
  13. Joseph NM, Morrison SJ. Toward an understanding of the physiological function of Mammalian stem cells. Dev Cell. 2005 Aug;9:173-83.
  14. Poliwoda S, Noor N, Downs E, Schaaf A, Cantwell A, Ganti L, et al. Stem cells: a comprehensive review of origins and emerging clinical roles in medical practice. Orthop Rev (Pavia). 2022 Aug 25;14:37498.
  15. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, et al. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002 Dec;13:4279-95.
  16. Kolios G, Moodley Y. Introduction to stem cells and regenerative medicine. Respiration. 2013;85:3-10.
  17. Çavuşoğlu T, Uyanıkgil Y, Root Application; Local and Current Clinical Applications of Mesenchymal. FNG & Bilim Tıp Transplantasyon Dergisi 2016 Oct;1:72-83
  18. Cartwright, Mark. "Prometheus." World History Encyclopedia. 2013 Apr 20.
  19. Yamashita YM, Mahowald AP, Perlin JR, Fuller MT. Asymmetric inheritance of mother versus daughter centrosome in stem cell division. Science. 2007 Jan 26;315:518-21.
  20. Gomes AC, Saraiva M, Gomes MS. The bone marrow hematopoietic niche and its adaptation to infection. Semin Cell Dev Biol. 2021 Apr;112:37-48.
  21. Sarvar DP, Effatpanah H, Akbarzadehlaleh P, Shamsasenjan K. Mesenchymal stromal cell-derived extracellular vesicles: a novel approach in hematopoietic stem cell transplantation. Stem Cell Res Ther. 2022 May 16;13:202.
  22. Pala HG, Pala EE, Artunc Ulkumen B, Aktug H, Yavasoglu A, Korkmaz HA, et al. The protective effect of granulocyte colony-stimulating factor on endometrium and ovary in a rat model of diabetes mellitus. Gynecol Obstet Invest. 2014;78:94-100.
  23. Beerman I, Maloney WJ, Weissmann IL, Rossi DJ. Stem cells and the aging hematopoietic system. Curr Opin Immunol. 2010 Aug;22:500-6.
  24. Hortu I, Ozceltik G, Sahin C, Akman L, Yildirim N, Erbas O. Granulocyte Colony-Stimulating Factor Prevents Ischemia/Reperfusion-Induced Ovarian Injury in Rats: Evaluation of Histological and Biochemical Parameters. Reprod Sci. 2019 Oct;26:1389-94.
  25. Fathi E, Ehsani A, Sanaat Z, Vandghanooni S, Farahzadi R, Montazersaheb S. Hematopoietic Stem Cells Characteristics: From Isolation to Transplantation. Curr Stem Cell Res Ther. 2022;17:407-14.
  26. Frisch BJ. Hematopoietic Stem Cell Cultures and Assays. Methods Mol Biol. 2021;2230:467-77.
  27. Elahi S. Hematopoietic responses to SARS-CoV-2 infection. Cell Mol Life Sci. 2022 Mar 13;79:187.
  28. Zhang S, Dong F, Liu ZX, Ema H. Hematopoietic Stem Cells Differentiate into the Megakaryocyte Lineage--Review. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 2020 Jun;28:1044-48.
  29. Noetzli LJ, French SL, Machlus KR. New Insights Into the Differentiation of Megakaryocytes From Hematopoietic Progenitors. Arterioscler Thromb Vasc Biol. 2019 Jul;39:1288-1300.
  30. Katayama Y. Guest editorial: hematopoietic regulators in the marrow: new players in inter-organ communication. Int J Hematol. 2014 Jun;99:677-8.
  31. Bertrand JY, Traver D. Hematopoietic cell development in the zebrafish embryo. Curr Opin Hematol. 2009 Jul;16:243-8.
  32. Brown G. The Social Norm of Hematopoietic Stem Cells and Dysregulation in Leukemia. Int J Mol Sci. 2022 May 3;23:5063.
  33. Medvinsky A, Rybtsov S, Taoudi S. Embryonic origin of the adult hematopoietic system: advances and questions. Development. 2011 Mar;138:1017-31.
  34. Li N, Chen H, Wang J. DNA damage and repair in the hematopoietic system. Acta Biochim Biophys Sin (Shanghai). 2022 Jan 25;54:847-57.
  35. Bigildeev AE, Petinati NA, Drize NJ. How Methods of Molecular Biology Shape Our Understanding of the Hematopoietic System. Mol Biol (Mosk). 2019 Sep-Oct;53:711-24.
  36. Zhang CX, Liu F. Regulatory signaling pathways in hematopoietic stem cell development. Yi Chuan. 2021 Apr 20;43:295-307.
  37. Orelio C, Dzierzak E. Bcl-2 expression and apoptosis in the regulation of hematopoietic stem cells. Leuk Lymphoma. 2007 Jan;48:16-24.
  38. Klump, H. Hematopoietic Stem Cells. In: Brand-Saberi, B. (eds) Essential Current Concepts in Stem Cell Biology. Learning Materials in Biosciences. Springer, Cham. 2020.
  39. Siapati EK, Roubelakis MG, Vassilopoulos G. Liver Regeneration by Hematopoietic Stem Cells: Have We Reached the End of the Road? Cells. 2022 Jul 27;11:2312.
  40. Schmidt A. Shining a light on hematopoietic stem cells. Elife. 2022 Aug 19;11:e81963.
  41. Wright DE, Bowman EP, Wagers AJ, Butcher EC, Weissman IL. Hematopoietic stem cells are uniquely selective in their migratory response to chemokines. J Exp Med. 2002 May 6;195:1145-54.
  42. Yamashita M, Iwama A. Aging and Clonal Behavior of Hematopoietic Stem Cells. Int J Mol Sci. 2022 Feb 9;23:1948.
  43. Aggarwal R, Lu J, Pompili VJ, Das H. Hematopoietic stem cells: transcriptional regulation, ex vivo expansion and clinical application. Curr Mol Med. 2012 Jan;12:34-49.
  44. Miao R, Chun H, Feng X, Gomes AC, Choi J, Pereira JP. Competition between hematopoietic stem and progenitor cells controls hematopoietic stem cell compartment size. Nat Commun. 2022 Aug 8;13:4611.
  45. Jingjing Li, Osmond Lao, Freya F. Bruveris, Liyuan Wang, Kajal Chaudry, et al. Mimicry of embryonic circulation enhances the hoax hemogenic niche and human blood development. Cell Reports, 2022;40:111339
  46. Huang X, Cho S, Spangrude GJ. Hematopoietic stem cells: generation and self-renewal. Cell Death Differ. 2007 Nov;14:1851-9.
  47. Keller G, Lacaud G, Robertson S. Development of the hematopoietic system in the mouse. Exp Hematol. 1999 May;27:777-87.
  48. Glauche I, Moore K, Thielecke L, Horn K, Loeffler M, Roeder I. Stem cell proliferation and quiescence--two sides of the same coin. PLoS Comput Biol. 2009 Jul;5:e1000447.
  49. Sever IH, Ozkul B, Bozkurt MF, Erbas O. Therapeutic effect of finasteride through its antiandrogenic and antioxidant role in a propionic acid-induced autism model: Demonstrated by behavioral tests, histological findings and MR spectroscopy. Neurosci Lett. 2022 May 14;779:136622.
  50. Jurecic R. Hematopoietic Stem Cell Heterogeneity. Adv Exp Med Biol. 2019;1169:195-211.
  51. Silva WN, Costa AC, Picoli CC, Rocha BGS, Santos GSP, Costa PAC, et al. Hematopoietic stem cell stretches and moves in their bone marrow niche. Crit Rev Oncol Hematol. 2021 Jul;163:103368.
  52. Rao I, Crisafulli L, Paulis M, Ficara F. Hematopoietic Cells from Pluripotent Stem Cells: Hope and Promise for the Treatment of Inherited Blood Disorders. Cells. 2022 Feb 5;11:557.
  53. Porteus MH, Pavel-Dinu M, Pai SY. A Curative DNA Code for Hematopoietic Defects: Novel Cell Therapies for Monogenic Diseases of the Blood and Immune System. Hematol Oncol Clin North Am. 2022 Aug;36:647-65.
  54. Gallagher PG. Extramedullary hematopoietic stem cells. Blood. 2022 Jun 9;139:3353-4.
  55. Hortu I, Ozceltik G, Karadadas E, Erbas O, Yigitturk G, Ulukus M. The Role of Ankaferd Blood Stopper and Oxytocin as Potential Therapeutic Agents in Endometriosis: A Rat Model. Curr Med Sci. 2020 Jun;40:556-62.
  56. Joshi A, Kundu M. Mitophagy in hematopoietic stem cells: the case for the exploration. Autophagy. 2013 Nov 1;9:1737-49.
  57. Tadokoro Y, Hirao A. The Role of Nutrients in Maintaining Hematopoietic Stem Cells and Healthy Hematopoiesis for Life. Int J Mol Sci. 2022 Jan 29;23:1574.
  58. Filippi MD, Ghaffari S. Mitochondria in the maintenance of hematopoietic stem cells: new perspectives and opportunities. Blood. 2019 May 2;133:1943-52.
  59. Xu Y, Murphy AJ, Fleetwood AJ. Hematopoietic Progenitors and the Bone Marrow Niche Shape the Inflammatory Response and Contribute to Chronic Disease. Int J Mol Sci. 2022 Feb 17;23:2234.
  60. Kristensen HB, Andersen TL, Patriarca A, Kallenbach K, MacDonald B, Sikjaer T, et al. Human hematopoietic microenvironments. PLoS One. 2021 Apr 20;16:e0250081.
  61. Woolthuis CM, de Haan G, Huls G. Aging of hematopoietic stem cells: Intrinsic changes or micro-environmental effects? Curr Opin Immunol. 2011 Aug;23:512-7.
  62. Urao N, Liu J, Takahashi K, Ganesh G. Hematopoietic Stem Cells in Wound Healing Response. Adv Wound Care (New Rochelle). 2022 Nov;11:598-621.
  63. Suda T. Hematopoietic factors and hematological diseases. Jpn J Med. 1991 Nov-Dec;30:600-2.
  64. Kiel MJ, Radice GL, Morrison SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell. 2007 Aug 16;1:204-17.
  65. Erbaş O, Altuntaş İ. Oxytocin and Neuroprotective Effects [Internet]. Oxytocin and Health. IntechOpen; 2021. Available from: http://dx.doi.org/10.5772/ intechopen.96527
  66. Böhm L, Helbing DL, Oraha N, Morrison H. The peripheral nervous system in hematopoietic stem cell aging. Mech Ageing Dev. 2020 Oct;191:111329.
  67. Morganti C, Ito K. Mitochondrial Contributions to Hematopoietic Stem Cell Aging. Int J Mol Sci. 2021 Oct 15;22:11117.
  68. Yamada T, Park CS, Lacorazza HD. Genetic control of quiescence in hematopoietic stem cells. Cell Cycle. 2013 Aug 1;12:2376-83.
  69. Kandarakov O, Belyavsky A, Semenova E. Bone Marrow Niches of Hematopoietic Stem and Progenitor Cells. Int J Mol Sci. 2022 Apr 18;23:4462.
  70. Arat M. Clinical use of hematopoietic stem cells. FNG & Bilim Tıp Transplantasyon Dergisi. 2016;1:10-18
  71. Lee-Thedieck C, Schertl P, Klein G. The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev. 2022 Feb;181:114069.
  72. Faisal M, Hassan M, Kumar A, Zubair M, Jamal M, Menghwar H, et al. Hematopoietic Stem and Progenitor Cells (HSPCs) and Hematopoietic Microenvironment: Molecular and Bioinformatic Studies of the Zebrafish Models. Int J Mol Sci. 2022 Jun 30;23:7285.