Colony stimulating factor 1 receptor

Source: Wikipedia, the free encyclopedia.
CSF1R
Gene ontology
Molecular function
Cellular component
Biological process
Sources:Amigo / QuickGO
Ensembl
UniProt
RefSeq (mRNA)

NM_001288705
NM_005211
NM_001349736
NM_001375320
NM_001375321

NM_001037859
NM_007779

RefSeq (protein)

NP_001275634
NP_005202
NP_001336665
NP_001362249
NP_001362250

NP_001032948

Location (UCSC)Chr 5: 150.05 – 150.11 MbChr 18: 61.23 – 61.27 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Colony stimulating factor 1 receptor (CSF1R), also known as macrophage colony-stimulating factor receptor (M-CSFR), and CD115 (Cluster of Differentiation 115), is a cell-surface

neurodegeneration, and inflammatory bone diseases
.

Gene

In the human genome, the CSF1R gene is located on chromosome 5 (5q32), and in mice the Csf1r gene is located on chromosome 18 (18D). CSF1R is 60.002 kilobases (kbs) in length.

ribosomal protein L7 processed pseudogene, oriented in the opposite direction to the CSF1R gene.[5]

Protein

Schematic showing how ligand binding to receptor tyrosine kinases (RTKs) such as CSF1R promotes receptor dimerization which is important for receptor signaling.

CSF1R, the protein encoded by the CSF1R gene is a

signaling cascades triggered upon binding to CSF1R.[8]

Table 1. CSF-1R signaling proteins in myeloid cells[9]
Protein Full protein name; function
SFK Src family tyrosine kinases
Grb2 Adaptor
Mona Monocyte adaptor; adaptor
Socs1
Suppressor of cytokine signaling-1; adaptor
PLCγ
Phospholipase C-γ
p85 PI3K Regulatory subunit of PI3K
Cbl Casitas B lineage; ubiquitin ligase, adaptor
FMIP
FMS-interacting protein; function unknown
PP2A
Protein phosphatase 2A; serine/threonine phosphatase
Pyk2
Proline-rich and Ca2C-activated tyrosine kinase
Paxilin Focal complex adaptor
PTPφ
PTP for phosphopaxillin
MAYP/PSTPIP2 Macrophage actin-associated and tyrosine-phosphorylated protein; actin bundling
Iba1
Ionized Ca2C-binding adaptor protein 1; actin bundling
Gab2 Grb2-associated binder-2; Adaptor
Gab3
Grb2-associated binder-3; adaptor
SHIP1
SH2-domain-containing polyinositol phosphatase-1
SHP1
SH2-domain-containing phosphatase-1; PTP
SHP2
SH2-domain-containing phosphatase-2; PTP
PKC-δ
Protein kinase C-d
Pkare
PKA-related gene (Pkare); protein kinase
MysPDZ
110-kDa myosin XVIIIA
STAT1, STAT3, STAT5 Signal transducers and activators of transcription-1, -2, -3; transcription factors
Dok1, Dok2, Dok3 Downstream of kinase-1, -2, -3; adaptors
Vav Rho family guanine-nucleotide-exchange factor
BLIMP-1
B-lymphocyte-induced maturation protein-1; transcriptional repressor

Function

Osteoclasts

Illustrated cross-section of an activated osteoclast.

Osteoclast are multi-nucleated cells that that absorb and remove bone which is critical for growth of new bones and maintenance of bone strength. Osteoclasts are critical for the bone remodeling cycle which is achieved by the building of bone by osteoblasts, reabsorption by osteoclasts, and remodeling by osteoblasts.[10] Osteoclasts precursor cells and mature osteoclast require stimulation of CSF1R for survival. Blockage of CSF1R signaling prevents osteoclast precursor cells from proliferating, maturing, and fusing into multi-nucleated cells. Stimulation of CSF1R promotes osteoclastogenesis (differentiation of monocytes into osteoclasts). CSF1R signaling in osteoclasts precursors promotes survival by upregulation of the Bcl-X(L) protein, an inhibitor of pro-apoptotic caspase-9. CSF1R signaling in mature osteoclasts promotes survival by stimulating mTOR/S6 kinase and the Na/HCO3 co-transporter, NBCn1.[11] CSF1R signaling also directly regulates osteoclast function. Osteoclasts migrate along the bone surface, then adhere to the bone to degrade and reabsorb the bone matrix. CSF1R signaling positively regulates this behavior, increasing osteoclast chemotaxis and bone reabsorption.[10]

Monocytes and macrophages

Illustrated schematic of monocytes, monocyte-derived macrophages, and liver-resident Kupffer cells.

Monocytes and macrophages are

PI3K signaling pathways.[9]

Microglia

Immunofluorescence staining of homeostatic microglia in a healthy adult mouse retina.

CD11b and CD18) to synapse-bound iC3b. Csf1r loss-of-function inhibits synaptic pruning and leads to excessive non-functional synapses in the brain. In adulthood, CSF1R is required for the proliferation and survival of microglia.[12] Inhibition of CSF1R signaling in adulthood causes near-complete (>99%) depletion (death) of brain microglia, however reversal of CSF1R inhibition stimulates remaining microglia to proliferate and repopulate microglia-free niches in the brain.[13] Production of CSF1R ligands CSF-1 and IL-34 is increased in the brain following injury or viral infection, which directs microglia to proliferate and execute immune responses.[12]

Neural progenitor cells

Immunofluorescence staining of neural progenitor cells (green), vasculature (red), and astrocytes (blue).

CSF1R signaling has been found to play important roles in non-myeloid cells such as neural progenitor cells,

multipotent cells that are able to self-renew or terminally differentiate into neurons, astrocytes and oligodendrocytes. Mice with Csf1r loss-of-function have a significantly more neural progenitor cells in generative zones and fewer matured neurons in forebrain laminae due to failure of progenitor cell maturation and radial migration. These phenotypes were also seen in animals with Csf1r conditional knock-out specifically in neural progenitor cells, suggesting that CSF1R signaling by neural progenitor cells is important for maturation of certain neurons.[11] Studies using cultured neural progenitor cells also show that CSF1R signaling stimulates neural progenitor cells maturation.[12]

Germline cells

CSF1R is expressed in oocytes, the trophoblast, and fertilized embryos prior to implantation in the uterus.[8] Studies using early mouse embryos in vitro have shown that activation of CSF1R stimulates formation of the blastocyst cavity and enhances the number of trophoblast cells. Csf1r loss-of-function mice exhibit several reproductive system abnormalities in the estrous cycle and ovulation rates as well as reduced antral follicles and ovarian macrophages. It is not clear whether ovulation dysfunction in Csf1r loss-of-function mice is due to loss of the protective effects of ovarian macrophages or loss of CSF1R signaling in oocytes themselves.[11]

Clinical significance

Bone disease

X-ray (A), CT scan (B) and MRI (C, D) show osteolysis of bone in the skull of an individual with Gorham-Stout disease.

endothelial cells similarly produces excessive osteoclastogenesis and osteolysis.[8] Additionally, postmenopausal loss of estrogen has also been found to impact CSF1R signaling and cause osteoporosis. Estrogen deficiency causes osteoporosis by upregulating production of TNF-α by activated T cells. As in inflammatory arthritis, TNF-α stimulates stromal cells to produce CSF-1 which increases CSF1R signaling in osteoclasts.[15]

Cancer

Schematic of tumor microenvironment of breast cancer. Cancers secrete many signals including colony stimulating factor which activates TAM CSF1R and promotes tumor growth and survival.

acute myeloblastic leukemia.[17]

Neurological disorders

Adult-onset leukoencephalopathy

Because of the importance of the CSF1R gene in myeloid cell survival, maturation, and function, loss-of-function in both inherited copies of the CSF1R gene causes postnatal mortality.

Nasu-Hakola disease (caused by mutations in either DAP12 or TREM2) and adult-onset leukoencephalopathy suggest partial loss of microglia CSF1R signaling promotes neurodegeneration. Defects in neurogenesis and neuronal survival are also seen in adult-onset leukoencephalopathy due to impaired CSF1R signaling in neural progenitor cells.[12]

Other brain diseases and disorders

Schematic of brain microglia and infiltrating peripheral macrophages mounting immune responses against β-amyloid plaque.

CSF1R signaling is involved in several diseases and disorders of the

Charcot-Marie-Tooth disease type 1, CSF-1 secretion from endoneurial cells stimulates proliferation and activation of macrophages and microglia that cause demyelination. Likewise in multiple sclerosis, CSF1R signaling supports the survival of inflammatory microglia which promote demyelination. CSF1R inhibition prophylactically reduces demyelination in the experimental autoimmune encephalomyelitis animal model. The role of CSF1R signaling in Alzheimer's disease is more complicated because microglia both protect and damage the brain in response to Alzheimer's disease pathology. CSF-1 stimulates primary cultured human microglia to phagocytose toxic 1–42 peptides. Microglia also initiate TREM2-dependent immune responses to amyloid plaques which protects neurons.[19][20] However, Alzheimer's disease microglia also excessively secrete inflammatory cytokines and prune synapses promoting synapse loss, neuronal death, and cognitive impairment.[21] Both CSF1R stimulation and inhibition improves cognitive function in Alzheimer's disease models.[12] Thus, microglia seem to have both protective and neurotoxic functions during Alzheimer's disease neurodegeneration.[22][23] Similar findings have been reported in lesion studies of the mouse brain, which showed that inhibition of CSF1R after lesioning improves recovery but inhibition during lesioning worsens recovery.[12]
CSF1R-targeting therapies for neurological disorders may impact both detrimental and beneficial microglia functions.

Therapeutics

Chemical structure of Pexidartinib (PLX3397), an FDA-approved small molecule inhibitor of CSF1R.
Structure of monoclonal antibody Emactuzumab light and heavy chain complexed with CSF1R.

Because

FDA-approved for treatment of diffuse-type tenosynovial giant cell tumors, a non-malignant tumor that develops from synovial tissue lining the joints.[24]

Table 2. CSF1R-targeted therapies in clinical development[8][16]
Drug name Form Targets Clinical trial diseases
Pexidartinib (PLX3397) Small molecule CSF1R,
Flt3
acral melanoma, mucosal melanoma
Imatinib Small molecule CSF1R,
PDGFR-β
chronic myeloid leukemia (CML), breast cancer
PLX5622 Small molecule CSF1R Rheumatoid arthritis, cancer, neuropathic pain, Alzheimer's disease
Sotuletinib (BLZ945) Small molecule CSF1R, c-KIT, PDGFRβ, and Flt3
amyotrophic lateral sclerosis
GW2580 Small molecule CSF1R Arthritis, osteoporosis, cancer
Ki20227 Small molecule CSF1R, VEGFR2, c-KIT, and PDGFRβ Osteolysis, breast cancer
Edicotinib

(JNJ-40346527)

Small molecule CSF1R, c-KIT, and Flt3 Alzheimer's disease, cHL, rheumatoid arthritis, neurodegenerative diseases
Emactuzumab (RG7155) Monoclonal antibody CSF1R Solid tumors
IMC-CS4 (LY3022855) Monoclonal antibody CSF1R Solid tumors, breast cancer, prostate cancer
AMG820 Monoclonal antibody CSF1R Solid tumors

Safety of CSF1R inhibition

The safety of CSF1R inhibitors has been extensively characterized in

lacrimation, and reduced appetite, but no signs of liver toxicity were found. There are some differences in the side effects of monoclonal antibody compared to small molecule CSF1R inhibitors. Edema was more common with monoclonal antibody treatment compared to small molecules, suggesting that immune response to monoclonal antibodies may drive some side effects. Additionally, some small molecule inhibitors are not specific for CSF1R, and off-target effects could explain observed side effects. For example, Pexidartinib treatment was found to change hair color, presumably by its impact on KIT kinase. Overall, CSF1R inhibitors have favorable safety profiles with limited toxicity.[16]

Controversy

CSF1R inhibitors such as PLX5622 are widely used to study the role of microglia in mouse

preclinical models of Alzheimer's disease, stroke, traumatic brain injury, and aging. PLX5622 is typically used for microglia research because PLX5622 has higher brain bioavailability and CSF1R-specificity compared to other CSF1R inhibitors such as PLX3397.[13] In 2020, researchers David Hume (University of Queensland) and Kim Green (UCI) published a letter in the academic journal PNAS defending the use small molecule CSF1R inhibitors to study microglia in brain disease.[25] This letter was in response to a primary research paper published in PNAS by lead correspondent Eleftherios Paschalis (HMS) and others which provided evidence that microglia research using PLX5622 is confounded by CSF1R inhibition in peripheral macrophages. Paschalis and colleagues published a subsequent letter in PNAS defending the findings of their published research.[26]

Interactions

Colony stimulating factor 1 receptor has been shown to

interact
with:

See also

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000182578Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000024621Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. ^ a b EntrezGene 1436
  6. PMID 1611909
    .
  7. ^ .
  8. ^ .
  9. ^ .
  10. ^ .
  11. ^ .
  12. ^ .
  13. ^ .
  14. .
  15. .
  16. ^ .
  17. .
  18. .
  19. .
  20. .
  21. .
  22. .
  23. .
  24. ^ Center for Drug Evaluation and Research (2019-12-20). "FDA approves pexidartinib for tenosynovial giant cell tumor". FDA.
  25. PMID 33446486
    .
  26. .
  27. .
  28. .
  29. .
  30. .

Further reading

External links

This article incorporates text from the United States National Library of Medicine, which is in the public domain.