HIF1A

Source: Wikipedia, the free encyclopedia.
HIF1A
Gene ontology
Molecular function
Cellular component
Biological process
Sources:Amigo / QuickGO
Ensembl
UniProt
RefSeq (mRNA)

NM_181054
NM_001243084
NM_001530

NM_010431
NM_001313919
NM_001313920

RefSeq (protein)

NP_001230013
NP_001521
NP_851397
NP_001521.1

NP_001300848
NP_001300849
NP_034561

Location (UCSC)Chr 14: 61.7 – 61.75 MbChr 12: 73.95 – 73.99 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

Hypoxia-inducible factor 1-alpha, also known as HIF-1-alpha, is a subunit of a heterodimeric

The Nobel Prize in Physiology or Medicine
2019 was awarded for the discovery of HIF.

HIF1A is a

vascularization and angiogenesis, energy metabolism, cell survival, and tumor invasion.[7][10] The presence of HIF1A in a hypoxic environment is required to push forward normal placental development in early gestation. [11]
Two other
isoforms have been identified.[7]

Structure

HIF1 is a

C-terminal, followed by two distinct PAS (PER-ARNT-SIM) domains, and a PAC (PAS-associated C-terminal) domain.[8][6] The HIF1A polypeptide also contains a nuclear localization signal motif, two transactivating domains CTAD and NTAD, and an intervening inhibitory domain (ID) that can repress the transcriptional activities of CTAD and NTAD.[13] There are a total of three HIF1A isoforms formed by alternative splicing, however isoform1 has been chosen as the canonical structure, and is the most extensively studied isoform in structure and function.[14][15]

Gene and expression

The human HIF1A gene encodes for the alpha subunit, HIF1A of the transcription factor hypoxia-inducible factor (HIF1).

hypoxia, HIF1A transcription is often significantly upregulated.[18][19][20][21][22][23] Typically, oxygen-independent pathway regulates protein expression, and oxygen-dependent pathway regulates degradation.[10] In hypoxia-independent ways, HIF1A expression may be upregulated through a redox-sensitive mechanism.[24]

Function

Nobel Prize in Physiology/ Medicine 2019: Cellular Oxygen Sensing and Adaption by Hif-alpha

The transcription factor HIF-1 plays an important role in cellular response to systemic oxygen levels in mammals.

VEGF and erythropoietin that are involved in biological processes such as angiogenesis and erythropoiesis, which assist in promoting and increasing oxygen delivery to hypoxic regions.[10][28][27] HIF-1 also induces transcription of genes involved in cell proliferation and survival, as well as glucose and iron metabolism.[27] In accordance with its dynamic biological role, HIF-1 responds to systemic oxygen levels by undergoing conformational changes, and associates with HRE regions of promoters of hypoxia-responsive genes to induce transcription.[29][30][31][32][33]

HIF1A stability, subcellular localization, as well as transcriptional activity are especially affected by oxygen level. The alpha subunit forms a heterodimer with the beta subunit. Under

tumor suppressor protein (VHL), which itself is part of a ubiquitin ligase enzyme.[37][38] Once the hydrolylated HIF1A is buried in the VHL protein, VHL will transport it to a proteasome to digest and destroy HIF1A. This prevents HIF1A from entering into the cell nucleus to carry out the transcription of many different regulatory pathways. Many of these pathways are necessary for proper placental development in early gestation. Under normoxic conditions the HIF1A will be hydroxylated and destroyed, which leads to placental tissue necrosis, disorganization, and overgrowth. [39][40]The hydroxylation of HIF1A proline residue also regulates its ability to associate with co-activators under hypoxia.[41][42] Function of HIF1A gene can be effectively examined by siRNA knockdown based on an independent validation.[43]

Repair, regeneration and rejuvenation

In normal circumstances after injury HIF1A is degraded by prolyl hydroxylases (PHDs). In June 2015, scientists found that the continued up-regulation of HIF1A via PHD inhibitors regenerates lost or damaged tissue in mammals that have a repair response; and the continued down-regulation of HIF1A results in healing with a scarring response in mammals with a previous regenerative response to the loss of tissue. The act of regulating HIF1A can either turn off, or turn on the key processes of mammalian regeneration.[44][45] One such regenerative process in which HIF1A is involved is peripheral nerve regeneration. Following axon injury, HIF1A activates VEGFA to promote regeneration and functional recovery.[46][47] HIF1A also controls skin healing.[48] Researchers at the Stanford University School of Medicine demonstrated that HIF1A activation was able to prevent and treat chronic wounds in diabetic and aged mice. Not only did the wounds in the mice heal more quickly, but the quality of the new skin was even better than the original.[49][50][51][52] Additionally the regenerative effect of HIF-1A modulation on aged skin cells was described[53][54] and a rejuvenating effect on aged facial skin was demonstrated in patients.[55] HIF modulation has also been linked to a beneficial effect on hair loss.[56] The biotech company Tomorrowlabs GmbH, founded in Vienna in 2016 by the physician Dominik Duscher and pharmacologist Dominik Thor, makes use of this mechanism.[57] Based on the patent-pending HSF ("HIF strengthening factor") active ingredient, products have been developed that are supposed to promote skin and hair regeneration.[58][59][60][61]

Regulation

HIF1A abundance (and its subsequent activity) is regulated transcriptionally in an

prolyl hydroxylases (PHDs) maintain the appropriate balance of HIF1A protein in the post-translation phase.[64]

PHDs rely on iron among other molecules to hydroxylate HIF1A; as such, iron chelators such as

desferrioxamine (DFO) have proven successful in HIF1A stabilization.[65] HBO (Hyperbaric oxygen therapy) and HIF1A imitators such as cobalt chloride have also been successfully utilized.[65]

Factors increasing HIF1A[66]

  • Modulator of Degradation:
  • Modulators of translation:
    • RNA-binding proteins,
      HuR
    • PtdIns3K and MAPK
      pathways
    • IRES-mediated translation
    • calcium signaling
    • miRNAs

Factors decreasing HIF1A[66]

Role in cancer

HIF1A is overexpressed in many human cancers.[67][68] HIF1A overexpression is heavily implicated in promoting tumor growth and metastasis through its role in initiating angiogenesis and regulating cellular metabolism to overcome hypoxia.[69] Hypoxia promotes apoptosis in both normal and tumor cells.[70] However, hypoxic conditions in tumor microenvironment especially, along with accumulation of genetic alternations often contribute to HIF1A overexpression.[10]

Significant HIF1A expression has been noted in most solid tumors studied, which include cancers of the

Studies of
gene transcription level.[79][80] In addition, high-grade glioblastoma multiform tumors with high VEGF expression pattern, similar to breast cancer with HIF1A overexpression, display significant signs of tumor neovascularization.[81] This further suggests the regulatory role of HIF1A in promoting tumor progression, likely through hypoxia-induced VEGF expression pathways.[80]

[71] HIF1A overexpression in tumors may also occur in a hypoxia-independent pathway. In hemangioblastoma, HIF1A expression is found in most cells sampled from the well-vascularized tumor.

AKT pathway is also involved in tumor growth. In prostate cancers, the commonly occurring PTEN mutation is associated with tumor progression toward aggressive stage, increased vascular density and angiogenesis.[83]

During hypoxia,

tumor suppressor p53 overexpression may be associated with HIF1A-dependent pathway to initiate apoptosis. Moreover, p53-independent pathway may also induce apoptosis through the Bcl-2 pathway.[70] However, overexpression of HIF1A is cancer- and individual-specific, and depends on the accompanying genetic alternations and levels of pro- and anti-apoptotic factors present. One study on epithelial ovarian cancer shows HIF1A and nonfunctional tumor suppressor p53 is correlated with low levels of tumor cell apoptosis and poor prognosis.[75] Further, early-stage esophageal cancer patients with demonstrated overexpression of HIF1 and absence of BCL2 expression also failed photodynamic therapy.[84]

While research efforts to develop therapeutic drugs to target hypoxia-associated tumor cells have been ongoing for many years, there has not yet been any breakthrough that has shown selectivity and effectiveness at targeting HIF1A pathways to decrease tumor progression and angiogenesis.

heterogenous
nature of the many cancer types and subtypes.

Interactions

HIF1A has been shown to

interact
with:

See also

  • Hypoxia inducible factors

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000100644Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000021109Ensembl, 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. PMID 8786149
    .
  6. ^ .
  7. ^ a b c "Entrez Gene: HIF1A hypoxia-inducible factor 1, alpha subunit (basic helix-loop-helix transcription factor)".
  8. ^
    PMID 7539918
    .
  9. .
  10. ^ .
  11. .
  12. .
  13. .
  14. .
  15. ^ "Hypoxia-inducible factor 1-alpha". 2014.
  16. ^ "HIF1A". National Center for Biotechnology Information.
  17. ^ "Anti-HIF1 alpha antibody (GTX127309) | GeneTex". www.genetex.com. Retrieved 2019-10-28.
  18. ^
    PMID 10425220
    .
  19. .
  20. .
  21. .
  22. .
  23. .
  24. .
  25. .
  26. .
  27. ^ .
  28. .
  29. .
  30. ^ .
  31. .
  32. .
  33. ^ .
  34. .
  35. .
  36. .
  37. .
  38. ^ .
  39. .
  40. .
  41. ^ .
  42. .
  43. .
  44. ^ eurekalert.org staff (3 June 2015). "Scientist at LIMR leads study demonstrating drug-induced tissue regeneration". eurekalert.org. Lankenau Institute for Medical Research (LIMR). Retrieved 3 July 2015.
  45. PMID 26041709
    .
  46. .
  47. .
  48. .
  49. ^ "Skin patch could help heal, prevent diabetic ulcers, study finds". Welcome to Bio-X. © Stanford University, Stanford, California 94305. 2015-01-23. Retrieved 2020-12-04.
  50. PMID 25535360
    .
  51. .
  52. .
  53. .
  54. .
  55. .
  56. .
  57. ^ Tomorrowlabs. "Tomorrowlabs". Tomorrowlabs. Retrieved 2020-12-04.
  58. ^ "Kosmetikbranche: Wie das Beauty-Start-up Tomorrowlabs den Markt erobert". www.handelsblatt.com (in German). Retrieved 2020-12-04.
  59. ^ "Ein Protein gegen das Altern und für das Geldverdienen". nachrichten.at (in German). Retrieved 2020-12-04.
  60. ^ "Das neue Beauty-Investment von Michael Pieper - HZ". Handelszeitung (in German). Retrieved 2020-12-04.
  61. ^ andrea.hodoschek (2020-08-03). "Milliardenmarkt Anti-Aging: Start-up aus Österreich mischt mit". kurier.at (in German). Retrieved 2020-12-04.
  62. PMID 18393939
    .
  63. ^ Rius, J., Guma, M., Schachtrup, C. et al. NF-κB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1α. Nature 453, 807–811 (2008). https://doi.org/10.1038/nature06905
  64. S2CID 2434206
    .
  65. ^ .
  66. ^ .
  67. ^ .
  68. ^ .
  69. ^ .
  70. ^ .
  71. ^ .
  72. .
  73. .
  74. .
  75. ^ .
  76. .
  77. .
  78. ^ .
  79. .
  80. ^ .
  81. .
  82. ^ .
  83. .
  84. .
  85. .
  86. .
  87. .
  88. .
  89. .
  90. ^ .
  91. .
  92. ^ .
  93. ^ .
  94. ^ .
  95. ^ .
  96. .
  97. .
  98. .
  99. ^ .
  100. ^ .
  101. .
  102. .
  103. .
  104. .
  105. .
  106. .
  107. .

Further reading

External links

This page is based on the copyrighted Wikipedia article: HIF1A. Articles is available under the CC BY-SA 3.0 license; additional terms may apply.Privacy Policy