Hematopoiesis: Stages, Sites, and Its Regulation

The combination of two Greek words, haima meaning blood and poiēsis meaning to produce something, forms the word hematopoiesis. So, hematopoiesis means forming blood cells (WBCs, RBCs, and platelets). 

Hematopoiesis begins in the embryonic stage and continues through adulthood to restore blood cells. It requires a multipotent hematopoietic stem cell to divide into progenitor cells. These progenitor cells can divide into specialized cells, forming mature white blood cells (WBCs), red blood cells (RBCs), and platelets.

Hematopoiesis, a cornerstone of our bodily functions, is a continuous process that tirelessly produces blood cells. Its significance lies in its role in ensuring the proper functioning of our immune system and the efficient transport of oxygen, making it a process we should truly appreciate. 

The location of hematopoiesis changes throughout life. It begins in the yolk sac and aorta-gonad-mesonephros. Then, it transitions into the liver, spleen, and finally, the bone marrow and lymph nodes. The process continues in these locations throughout adult life except in pathological conditions, when it can return to its former sites.  

Hematopoietic Microenvironment

The process of blood cell formation requires regulation by a specialized microenvironment known as the hematopoietic microenvironment or niche. The microenvironment comprises different types of adherent cells, such as fibroblasts, macrophages, and growth factors.

Components of Hematopoietic Microenvironment

The components of the hematopoietic microenvironment include bone marrow, cytokines, and growth factors, which interact to support hematopoiesis. 

  1. Bone marrow stromal cells: Fibroblasts, adipocytes, endothelial cells, and osteoblasts are some of the stromal cells involved in hematopoiesis. These provide structural support and secrete cytokines and growth factors required for hematopoiesis. 
  2. Growth factors and Cytokines: These include soluble factors like stem cell factor (SCF), interleukins (IL-3, IL-6), and granulocyte colony-stimulating factor (G-CSF). These help the survival, proliferation, and differentiation of HSCs and progenitor cells. 
  3. Extracellular matrix (ECM): This provides an area for cells to attach and migrate. It comprises proteins like collagen, fibronectin, and laminin. These can bind to the integrins present in hematopoietic stem cells to regulate their behavior.  

Hematopoietic Stem Cells

These specialized immature stem cells can mature or develop into different kinds of blood cells. They can also produce identical copies of themselves, which contributes to their purpose of providing a continuous supply of blood cells. 

Many HSCs possess a unique ability to remain in a dormant phase or quiescence state, a protective mechanism that shields them from exhaustion and damage. This adaptability allows HSCs to respond to various signals, aiding the body’s defense by increasing blood cell production during injury or infection.  

HScs were once thought to originate from the yolk sac, but they originate from the ventral endothelial wall of the embryonic aorta. They are present in the peripheral blood and the bone marrow. 

Examples of Hematopoietic Microenvironment Interactions

  • Leukemia: In leukemic conditions, malignant cells can alter the bone marrow microenvironment, disrupting normal hematopoiesis. For instance, leukemic cells may secrete factors that suppress normal HSC function or alter stromal cell behavior.
  • Bone Marrow Transplantation: Successful bone marrow transplants require the establishment of a supportive microenvironment for donor HSCs. Conditioning regimens that prepare the recipient’s marrow space and use growth factors post-transplant are essential for engraftment.

Stages of Hematopoiesis

There are different stages of hematopoiesis. Hematopoiesis begins with hematopoietic stem cells (HSCs), which either self-renew or differentiate into two different lineages. 

Once the HSCs choose one lineage, it stays committed and matures into the designated mature blood cells. So, the starting step of hematopoiesis is the self-renewal of HSCs, followed by their differentiation into a particular lineage.  

Self Renewal of Hematopoietic Stem Cells (HSCs)

The HSCs have the option to either self-renew or differentiate into mature blood cells. Two contrasting models are proposed to explain this choice. In the stochastic model, the decision is random, while in the instructive model, specific microenvironmental factors and cytokines influence the choice. It’s important to note that these models are not without their controversies, adding to the intrigue of HSC biology. 

Likewise, there are two types of HSCs: long-term and short-term HSCs. The primary function of long-term HSCs is self-renewal, and short-term HSCs are to differentiate into multipotent progenitors. 

HSCs differentiation and commitment to lineages

Short-term HSCs give rise to different lineages and single and multiple lineage progenitors. The multilineage progenitors have two main progenitors: common lymphoid progenitors (CLPs) and common myeloid progenitors (CMPs), which produce different blood cells. 

Upon differentiation, HSCs commit to a specific lineage and mature into respective blood cells. The common myeloid progenitors play a vital role in producing RBCs, platelets, basophils, eosinophils, neutrophils, or macrophages. Similarly, the common lymphoid progenitors contribute to the formation of T, B , or NK (natural killer) cells. The identification of stem cells, progenitors, and mature blood cells of different lineages is aided by distinct cell surface markers or CDs.

Maturation and Development of Progenitors 

Following different processes, the common myeloid and common lymphoid progenitors mature and develop into different blood cells, following different hematopoiesis processes. 

The diverse processes of hematopoiesis include myelopoiesis, lymphopoiesis, thrombopoiesis, and erythropoiesis, each leading to the production of specific blood cell types. 

  1. Myleopoiesis forms granulocytes (basophils, eosinophils, or neutrophils) or agranulocytes (macrophages and monocytes).
  2. Lymphopoiesis produces T and B lymphocytes.
  3. Erythropoiesis forms red blood cells or erythrocytes. 
  4. Thrombopoiesis forms platelets.

Regulation of Hematopoiesis

The regulation of hematopoiesis is essential for ensuring blood cells’ balanced and continuous production. Blood cells are a significant part of different bodily functions, including but not limited to the immune system. Various intrinsic and extrinsic methods are applied to tighten the regulation of hematopoiesis. 

Intrinsic Regulators

  1. Transcription Factors:
    • GATA-1: Essential for erythroid and megakaryocyte differentiation.
    • PU.1: Crucial for myeloid and lymphoid lineage differentiation.
    • RUNX1: It plays a key role in regulating HSCs and developing all hematopoietic lineages.
  2. Epigenetic Modifications:
    • DNA Methylation: Regulates gene expression by adding methyl groups to DNA, thereby influencing hematopoietic lineage commitment.
    • Histone Modifications: Acetylation, methylation, and phosphorylation of histones affect chromatin structure and gene expression, impacting hematopoietic differentiation.
  3. MicroRNAs: These are small non-coding RNAs that post-transcriptionally regulate gene expression, impacting various aspects of HSC maintenance, self-renewal, and differentiation.

Extrinsic Regulators

  1. Cytokines and Growth Factors:
    • Stem Cell Factor (SCF) binds to the c-Kit receptor on HSCs, promoting their survival and proliferation.
    • Erythropoietin (EPO) stimulates red blood cell production in response to hypoxia.
    • Granulocyte Colony-Stimulating Factor (G-CSF): This promotes the proliferation and differentiation of granulocyte precursors.
    • Thrombopoietin (TPO): Regulates platelet production by stimulating megakaryocyte development.
  2. Bone Marrow Microenvironment (Niche):
    • Endosteal Niche: It is rich in osteoblasts, maintains HSC quiescence, and supports self-renewal.
    • Vascular Niche: It comprises endothelial and perivascular cells, facilitating HSC activation and mobilization.
  3. Cell-Cell Interactions: Direct contact with stromal cells, osteoblasts, and endothelial cells via adhesion molecules (e.g., integrins, cadherins) is crucial for HSC maintenance and regulation.
  4. Paracrine and Juxtacrine Signaling:
    • Notch Signaling: Direct cell-cell interactions involving Notch receptors and ligands regulate HSC fate decisions.
    • Wnt Signaling: It is involved in the regulation of HSC self-renewal and differentiation through paracrine mechanisms.
  5. Extracellular Matrix (ECM): It provides structural support and presents biochemical signals to HSCs through components like collagen, fibronectin, and laminin.

Systemic Factors

  1. Hormones:
    • Glucocorticoids: This hormone influence the proliferation and differentiation of specific hematopoietic lineages.
    • Thyroid Hormones: Affect erythropoiesis and overall hematopoietic activity.
  2. Nutritional Status: Adequate levels of vitamins (e.g., B12, folate), iron, and other nutrients are essential for effective hematopoiesis.
  3. Immune Signals: Cytokines released during infection or inflammation (e.g., interleukins, interferons) can alter hematopoietic activity to meet increased demand for immune cells.

Feedback Mechanisms

  1. Negative Feedback:
    • EPO Production: It is regulated by oxygen levels; increased red blood cell production reduces hypoxia, decreasing EPO synthesis.
    • Immune Cell Regulation: High levels of mature immune cells can inhibit further production through feedback mechanisms involving cytokines.
  2. Homeostatic Balance: Constant monitoring and adjustment of blood cell production ensure that the levels of different blood cell types remain within physiological ranges.

Sites of Hematopoiesis

The site of hematopoiesis depends on the life stage. It begins in the embryonic stage and continues until the organism is alive. The site differs between the embryonic and adult stages.   

Embryonic Hematopoiesis

The first phase of embryonic hematopoiesis, known as primitive hematopoiesis, is a critical stage. It takes place in the extraembryonic yolk sac, where primitive erythrocytes are formed. These erythrocytes play a vital role in the embryo’s survival by facilitating oxygen transportation. Additionally, myeloid cells produced in the yolk sac develop into microglia and Langerhans cells, which migrate to the central nervous system. In humans, this phase commences after the 19th day of conception and extends until the 8th week. 

The second phase is intraembryonic hematopoiesis, which occurs in the dorsal aorta (aorta-gonad-mesonephros or AGM). Here, mesoderm derivatives develop into progenitor hematopoietic stem cells. This phase only occurs once in the entire lifetime, and these cells convert into CD 45 and major histocompatibility complex (MHC II) in adult life.    

Approximately five weeks into development, hematopoiesis undergoes a significant transition. It shifts from the yolk sac or AGM to the liver, which becomes a crucial site for the differentiation and expansion of progenitor HSCs. In some cases, these cells may also migrate to the spleen to differentiate into myeloid and lymphoid lineages.

At about 16-20 weeks of gestation, hematopoiesis can start occurring in the bone marrow, termed medullary hematopoiesis. This hematopoiesis continues after birth to adult life. 

Adult Hematopoiesis

The main areas for adult hematopoiesis are the skull, pelvic bones, vertebrae, and the metaphyseal region of long bones. The shaft of long bones can be replaced by adipose tissue. There is a high demand for red blood cells during neonatal and childhood life. This is why the active bone marrow of children is more significant than that of adults. Due to various disorders, the hematopoiesis can change back to its embryonic sites. 

Disorders related to Hematopoiesis

Disruptions in hematopoiesis, the blood cell-producing process, can lead to various hematological disorders. These disorders may arise due to genetic mutations, environmental factors, or diseases affecting bone marrow and hematopoietic stem cells (HSCs).

Anemias

  • Aplastic Anemia: Damage to the bone marrow can lead to a deficiency in all types of blood cells (pancytopenia). Its symptoms include fatigue, pallor, infections, and bleeding. The treatment includes immunosuppressive therapy, bone marrow transplants, and blood transfusions.
  • Iron-Deficiency Anemia: Insufficient iron, leading to reduced hemoglobin production, causes this type of anemia. The symptoms include fatigue, weakness, shortness of breath, and pallor. Iron supplements and dietary changes can be possible treatments.
  • Megaloblastic Anemia: Vitamin B12 or folate deficiency, leading to abnormal red blood cell production. Fatigue, pallor, shortness of breath, and neurological symptoms in B12 deficiency are some of its symptoms. The treatment can be vitamin B12 or folate supplements.

Leukemias

  • Acute Myeloid Leukemia (AML): Clonal proliferation of myeloid precursors leads to immature cells (blasts) accumulating in the bone marrow. Fatigue, fever, infections, and bleeding are the symptoms. Chemotherapy, targeted therapy, and bone marrow transplant can be possible treatments.
  • Acute Lymphoblastic Leukemia (ALL): Clonal proliferation of lymphoid precursors causes ALL. Its symptoms are fatigue, fever, infections, bleeding, and bone pain. Chemotherapy, targeted therapy, and bone marrow transplants are available treatments.
  • Chronic Myeloid Leukemia (CML): BCR-ABL fusion gene due to a chromosomal translocation (Philadelphia chromosome) is the reason behind CML. Symptoms include fatigue, weight loss, splenomegaly, and night sweats. Treatment can be the use of tyrosine kinase inhibitors (e.g., imatinib), chemotherapy, and bone marrow transplant.
  • Chronic Lymphocytic Leukemia (CLL): Clonal proliferation of mature B lymphocytes causes CLL. It is often asymptomatic initially; later symptoms include fatigue, weight loss, and lymphadenopathy. Treatment includes observation in the early stages, chemotherapy, and targeted therapies.

Myelodysplastic Syndromes (MDS) 

Clonal hematopoietic disorders characterized by ineffective hematopoiesis and a risk of progression to AML. Symptoms include anemia, infections, and bleeding due to cytopenias. Supportive care (e.g., transfusions), growth factors, hypomethylating agents, and stem cell transplants are possible treatments.

Myeloproliferative Neoplasms (MPNs)

  • Polycythemia Vera (PV): Excessive production of red blood cells due to JAK2 mutation. Its symptoms are headaches, dizziness, pruritus, and a risk of thrombosis. The treatment includes phlebotomy, low-dose aspirin, and JAK2 inhibitors.
  • Essential Thrombocythemia (ET): Overproduction of platelets, often due to JAK2 or MPL mutations, causes ET. Symptoms include thrombosis, bleeding, and headache. Low-dose aspirin and cytoreductive therapy are the possible treatments.
  • Primary Myelofibrosis (PMF): Fibrosis of the bone marrow, leading to pancytopenia and extramedullary hematopoiesis can cause PMF. Fatigue, weight loss, splenomegaly, and anemia are the symptoms. JAK2 inhibitors, supportive care, and stem cell transplants are possible treatments.

Lymphomas

  • Hodgkin Lymphoma: Malignant transformation of B cells, characterized by the presence of Reed-Sternberg cells, causes Hodgkin lymphoma. Painless lymphadenopathy, fever, night sweats, and weight loss are observed symptoms. Chemotherapy, radiation therapy, and stem cell transplant are available treatments.
  • Non-Hodgkin Lymphoma: A diverse group of lymphoid malignancies affects B, T, or NK cells. The symptoms include lymphadenopathy, fever, night sweats, and weight loss. Treatments include chemotherapy, radiation therapy, targeted therapies, and stem cell transplants.

Bone Marrow Failure Syndromes

  • Fanconi Anemia: Genetic disorder causing bone marrow failure, leading to pancytopenia. Symptoms observed are growth retardation, congenital anomalies, and increased cancer risk. Hematopoietic stem cell transplant and supportive care are the treatments available.
  • Diamond-Blackfan Anemia: Congenital erythroid aplasia leading to anemia. Symptoms include severe anemia in infancy and physical abnormalities. Corticosteroids, red blood cell transfusions, and stem cell transplants are the available treatments.

References

  1. Jagannathan-Bogdan, M., & Zon, L. I. (2013). Hematopoiesis. Development (Cambridge, England), 140(12), 2463–2467. https://doi.org/10.1242/dev.083147 
  2. Chapman J, Zhang Y. Histology, Hematopoiesis. [Updated 2023 May 1]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK534246/ 
  3. Morrison, S. J., Uchida, N., & Weissman, I. L. (1995). The biology of hematopoietic stem cells. Annual review of cell and developmental biology, 11, 35–71. https://doi.org/10.1146/annurev.cb.11.110195.000343
  4. Greenberger J. S. (1991). The hematopoietic microenvironment. Critical reviews in oncology/hematology, 11(1), 65–84. https://doi.org/10.1016/1040-8428(91)90018-8 
  5. Gaballa, M., & Ramos, C. A. (2019). Overview of normal hematopoiesis. Handbook of Benign Hematology. https://doi.org/10.1891/9780826149879.0001

Ashma Shrestha

Hello, I am Ashma Shrestha. I had recently completed my Masters degree in Medical Microbiology. Passionate about writing and blogging. Key interest in virology and molecular biology.

We love to get your feedback. Share your queries or comments

This site uses Akismet to reduce spam. Learn how your comment data is processed.

Recent Posts