HPG80

Last updated

hPG80 refers to the extracellular and oncogenic version of progastrin. This name first appeared in a scientific publication in January 2020. [1] Until that date, scientific publications only mention 'progastrin', without necessarily explicitly specifying whether it is intracellular (in the context of digestion) or extracellular (circulating and detectable in plasma) in the tumor pathological setting.

Contents

For more clarity, the remainder of this article uses exclusively the name hPG80 to refer to extracellular progastrin.

A link between this protein and cancer has been known for more than 30 years. hPG80 is involved in most of the biological functions that ensure the existence of cancer. [2] The peptide is secreted by tumor cells and found in the plasma of cancer patients from early stages. [1] It then has functions that are independent of digestion and totally different from progastrin and its only role as an intracellular precursor of gastrin.

Terminology

In the name hPG80, the "h" describes the human species: human; "PG" is a common script for the progastrin protein and the number 80 corresponds to the size of the protein: 80 amino acids.

The name hPG80 was thus proposed in the publication resulting from the work of Professor Benoît You under the management of Dominique Joubert and Alexandre Prieur in order to remove ambiguities between the intracellular version of the protein (in the function of digestion) and its extracellular version of the protein (in the case of cancer patients) which is no longer, despite its name, the precursor of gastrin. [1]  

Moreover, the existence of a phonetically identical peptide, the Pro-Gastrin Releasing Peptide (proGRP), accentuated a possible confusion around the name progastrin and the need for a specific name.

Chronology of scientific discoveries on hPG80

1990: hPG80 is secreted by pancreatic cancer cells. [3]

1993 - 1994: hPG80 is secreted by ovarian and colon cancer cells. [4] [5]

1996 - 1997: hPG80 is identified as necessary for the proliferation and tumorigenesis of colon cancer cells. [6]

2000 - 2001: The GAST gene is a target of the β-catenin/Tcf4 pathway. [7] The presence of hPG80 is demonstrated in the plasma of colorectal cancer patients. [8]

2003: hPG80 is involved in the dissociation regulation of adherent and tight junctions. [2] Src mediates the effects of hPG80 on cell growth. [9]

2005: Ras and β-catenin/Tcf4 pathways induce synergistic activation of the GAST gene, contributing to possible neoplastic progression. [10]

2007: Inhibition of hPG80 expression inhibits the Wnt/β-catenin pathway inducing decreased growth and tumor differentiation in intestinal tumor models. [11]

2012: p53 gene mutation increases hPG80-dependent colonic proliferation and cancer formation. [12]

2014: hPG80 is identified as a new pro-angiogenic factor. [13]

2016: Autocrine secretion of hPG80 promotes survival and self-renewal of stem cells in colon cancer. [14]

2017: The use of antibodies directed against hPG80 to target the Wnt pathway and cancer stem cells represents a new therapeutic track for cancer. [15]

2020: hPG80 is detected in the plasma of patients with 11 different types of cancer. An association between longitudinal variations in hPG80 levels and the efficacy of anti-cancer treatments has been demonstrated. [1]

Role in cancer

Through a variety of mechanisms, all crucial for tumor growth and survival, research has demonstrated the major role that hPG80 plays in tumor initiation and progression, generally using colorectal cancer as a model. Bardram was the first to hypothesize the presence of hPG80 in the early stages of the disease. [3] He evaluated the presence of hPG80 and its maturation products in the serum of patients with Zollinger-Ellison syndrome. This work showed that hPG80 measurement more accurately reflected tumor than the conventional measurement of amide gastrin. Indeed, hPG80 is more than 700 times more abundant than amide gastrin in colorectal tumors. [16]

In the early 90s, it was shown that hPG80 was not fully matured in human colon cancer cell lines and, more importantly, that it was secreted by in vitro cells. [5] These observations led to the study of autocrine and paracrine function of hPG80 in tumor cells. [17] Generally, it has been shown that hPG80 does not mature properly in cancer cells, particularly in colorectal cancer, because maturation enzymes are either absent or non-functional. [4] [5] [16] [18] [19] [20] [21]

In 1996, it was demonstrated that GAST gene expression is required for human colon cancer cell tumorigenesis. [6] 60 to 80% of colon cancers express the GAST gene.

Through different experimental configurations, alteration of the GAST gene or neutralization of hPG80 resulted in a decrease in the number of tumors in mice with a tumor predisposition (mutation on the APC gene). Conversely, a significant increase in tumor formation was observed in mice overexpressing progastrin and treated with azoxymethane (AOM), a chemical carcinogen. [7] [11] [15] [22] [23]

Pro-angiogenic factor

When a tumor develops, its need for oxygen and nutrients causes creation of new blood vessels. This process is called neo-angiogenesis. In 2015, hPG80 was shown to be a pro-angiogenic factor. [13] Indeed, hPG80 stimulates endothelial cells proliferation and migration and increases their ability to form capillary-like structures in vitro. Blocking the production of hPG80 (with shRNA) in xenograft cells in nude mice reduces tumor neovascularization.

Anti-apoptotic factor

Treatment of intestinal epithelial cells with hPG80 results in a significant loss of caspase 9 and caspase 3 activation and a decrease in DNA fragmentation. Therefore, the effect of hPG80 on cell survival results from both increased proliferation and decreased apoptosis. [24]

Regulation of cell junctions

Cell migration is based on their ability to become independent of adjacent cells. Intercellular contacts integrity is essential for electrolyte uptake regulation as well as for tumor metastasis prevention. In 2003, it was demonstrated that blocking hPG80 secretion by an antisense construction directed against progastrin mRNA allows restoration of membrane localization of tight and adherent junctions constituent proteins in a human colorectal carcinoma cell line DLD-1. [2] hPG80 thus plays a role on cell contacts integrity.

Role in cancer stem cells

Figure 2 - hPG80, Cornerstone in ongenetic pathways Capture d'ecran 2020-06-23 a 20.23.55.png
Figure 2 - hPG80, Cornerstone in ongenetic pathways

Cancer stem cells (CSCs) constitute a small proportion of the tumor, usually between 1 and 5%. But they are essential for tumor survival because they act as a "reactor". Giraud and al. have demonstrated the major role played by hPG80 in CSCs. [14] These authors have shown that hPG80 expression is strongly increased in colorectal cancer cells cultured under conditions where CSCs are enriched. They then showed that hPG80 was able to regulate CSCs frequency (survival and self-renewal) in vitro and in vivo. Subsequently, Prieur and al. demonstrated that when a neutralizing antibody is added to a cell culture or when human colorectal cancer cells implanted mice are treated in vivo with such an antibody, CSCs frequency is decreased in the same proportions. [15] These two scientific articles clearly show that hPG80 is a survival factor for CSCs. Moreover, hPG80 inhibition would induce CSC differentiation, opening the possibility of a differentiation therapy rather than a classical anti-proliferative therapy. Its main function is to help CSCs survive and spread to form metastases, which probably explains why this peptide can be considered as a potential predictive marker for the presence of liver metastasis in colorectal cancer. [25]

hPG80 and oncogenic signaling pathways

Wnt and K-Ras

The first event leading to colon cancer is in 80 to 90% of cases a constitutive activation of the Wnt/β-catenin oncogenic pathway induced by mutations in the APC (Adenomatous polyposis coli) coding gene or the β-catenin coding gene. Mutations induction into normal intestinal stem cells is sufficient to trigger tumorigenesis. [26] GAST gene which encodes hPG80 is activated by the Wnt oncogenic pathway. It is a downstream target of the β-catenin/Tcf-4 signaling pathway. [7]

Demonstration of the link between hPG80 and an oncogenic pathway has been described for K-Ras. Cell lines and colon cancer tissues with K-Ras mutations all had significantly higher levels of GAST mRNA than wild type K-Ras. [27] K-Ras effects on GAST expression are produced by activation of the Raf-MEK-ERK signal transduction pathway, final step being activation of the GAST gene promoter.

Since both K-Ras and the Wnt pathway induce GAST expression, the hypothesis of a possible cooperation between these two pathways to regulate hPG80 expression was investigated. [10] Chakadar and al. found significant synergistic stimulation of the GAST gene promoter (by a factor of 25-40) by a combination of oncogenic β-catenin and K-Ras overexpression. Activation of the GAST gene promoter was also shown to be dependent on other signalling signals: enhanced or suppressed by co-expression of wild-type SMAD4 or by a dominant negative mutant of SMAD4, respectively, and abrogated by inhibition of PI3K. Thus, constitutive activation of the Wnt pathway, considered to be at the onset of tumorigenesis in the colon, and the K-Ras oncogene, present in 50% of human colorectal tumors, synergistically stimulate the production of hPG80, a tumorigenesis promoter. Tumorigenesis induced by activation of the Wnt pathway is partially dependent on hPG80.

SRC, PI3K/Akt, NF-kB and Jak/Stat

The oncogene pp60 (c-Src) is activated in colon cancer cells and leads to increased amounts of hPG80. [9] This means that hPG80 production, which occurs during early tumorigenesis, could play a role in this activation, which is known as an early event in colon tumorigenesis. [11] [28] [29] The PI3K/Akt pathway, which is particularly involved in proliferation, is also activated by hPG80. NF-kappaB is another important signaling messenger regulated by hPG80. [11] [30] Its involvement in mechanisms responsible for the anti-apoptotic effect of hPG80 has been demonstrated in pancreatic cancer cells in vitro and in vivo in mice overexpressing the GAST gene. [31] [32] JAK2 (Janus-activated kinase 2), STAT3 and kinases increases regulated by extracellular signals have also been observed in the colon mucosa of hGAS mice. [30]

NOTCH

It has been shown that colon tumor cells which do not express hPG80 return to a "normal" state. This is due to the fact that when the Wnt pathway is inactivated, the JAG1 gene is repressed, inducing inactivation of the Notch pathway which plays a major role in the acquisition of a differentiated cell phenotype. [33]

p-53

In 2012, the P53 gene mutation was shown to increase hPG80-dependent colon cells proliferation and colon cancer formation in mice. [12]

hPG80 receptor

An identification of the hPG80 receptor has been the subject of several studies in recent years. However, hPG80 receptor identity remains a real issue in the scientific community. The receptor can activate a number of signaling pathways, either directly or indirectly, which is rather unusual for a receptor. This could indicate a peculiarity of this receptor and why it is difficult to identify it. The unidentified hPG80 receptor transduces a progastrin signal via various intracellular intermediates known to be involved in tumorigenesis.

High affinity binding sites have been described for the first time in intestinal epithelial cells using recombinant human progastrin iodine. [34] Affinity was in the range of 0.5-1 nM, which is receptor compatible. When biotinylated progastrin binding was evaluated by flow cytometry, strong and specific binding of progastrin to certain cell lines (IEC-6, IEC-18, HT-29, COLO320) was also detected. [35] A specificity of binding was confirmed by competition with cold, unlabelled hPG80, but not with carboxy-terminal glycine-containing gastrin or amidated gastrin-17. Binding was not influenced by the presence of the classical CCK-2 receptor.

It is clear from these two studies that there is a binding site/receptor for hPG80 that is distinct from binding to gastrin-17 amidated and to gastrin 17 with carboxy-terminal glycine (G17-Gly). The sequence of hPG80 interacting with this receptor is likely located in the last 26 amino acid residues of hPG80 carboxy-terminal end, which have been shown to be sufficient for its function. [36] However, this putative receptor identity is unknown.

One candidate is Annexin A2, identified as being able of binding progastrin and derived peptides. [37] Annexin A2 is a partial mediator of the effect of progastrin/gastrins. In particular, Annexin A2 mediates NF-kappaB upregulation and β-catenin in response to progastrin in mice and HEK-293 cells. [38] In addition, annexin A2 may be involved in progastrin-dependent clathrin endocytosis. However, progastrin affinity for Annexin A2 is not what would be expected for a specific receptor. And, although Annexin A2 plays a role in progastrin functions, it does not fit a receptor function.

Another candidate is the G protein-coupled receptor 56 (GPCR56), expressed on both colon stem cells and cancer cells. [39] While human recombinant hPG80 promotes in vitro growth and survival of wild mice colon organoids, those from GPCR56-deficient mice (GPCR56-/-) are resistant to hPG80. However, although hPG80 has been shown to bind to cells expressing GPCR56, authors did not provide direct evidence of a hPG80 bound to GPCR56 itself. GPCR56 is a good candidate, but evidence that it is hPG80 receptor is to this day not established.

Clinical Applications

hPG80 through various mechanisms can be considered as a major promoter of tumorigenesis. hPG80 is found in the plasma of cancer patients and its neutralization induces tumor reversion.

Screening

Universal biomarker

Colorectal cancer is not the only type of cancer to express hPG80. Its expression has also been demonstrated in ovarian cancers, liver tumors and pancreatic tumors. [4] [40] [41] Thus, several types of tumors express the unmatured peptide. hPG80 can be detected and quantified in the blood of cancer patients. A first demonstration has been made in colorectal cancer showing an increase of hPG80 and non-amide gastrin levels in plasma in these patients compared to a control series (healthy individuals). [8] In addition, an hPG80 increase has been observed in patients with adenomatous polyps. [15] hPG80 is therefore expressed at all stages by the tumor, from early stages to metastasis.

Multi-cancer detection

Figure 3 - Longitudinal changes in hPG80 and AFP levels during hepatocellular carcinoma management (inclusion; remission; progression) in a representative patient Variations des taux de hPG80 et AFP.png
Figure 3 - Longitudinal changes in hPG80 and AFP levels during hepatocellular carcinoma management (inclusion; remission; progression) in a representative patient

hPG80 is found in different tumor types and secreted in vitro by certain cancer cells. A study has shown its presence in the blood of patients with 11 different types of cancer: breast cancer, colorectal cancer, stomach/esophageal cancer, kidney cancer, liver cancer, non-small cell lung cancer, skin melanoma, ovarian cancer, pancreatic cancer, prostate cancer and uterine cancer (endometrial/cervical). [1]

hPG80 in residual disease surveillance and response to therapy.

Pre- and post-operative plasma assays of hPG80 in colorectal cancer patients show that hPG80 presence reflects tumor production. [42]

It has been observed that hPG80 concentrations are increased in patients at risk of developing colorectal carcinoma. [43] In addition, an increase in hPG80 has been observed in hyperplastic polyps that have progressed to cancer. [44] [45]

hPG80 may also be a biomarker of liver metastasis in colorectal cancer. [25]

In addition, You and al. observed a decrease in hPG80 levels after surgery in a patients cohort with gastrointestinal cancers with peritoneal involvement treated with post-operative chemotherapy and cytoreductive surgery. [1]

In addition, earlier detection of small lesions and monitoring of recurrence can be improved by measuring hPG80 levels as a complementary blood biomarker in a cohort of patients with hepatocellular carcinoma treated with local or systemic therapies (Fig. 3). [1] Measured hPG80 levels are relevant in patients for whom alpha-fetoprotein (AFP) is below 20 ng/ml, an established threshold in clinical practice. [46] Depending on disease management, patients in remission have lower levels of hPG80 than those in whom the cancer is still active.

Target to fight cancer

Antibody therapy

To date, no drug is able to target CSCs. Humanized anti-hPG80 antibodies, alone or in combination with chemotherapy, appears to be a promising approach. [15] The use of anti-hPG80 antibodies has also been discussed as a potential treatment in colorectal cancer patients with a mutation in the KRAS gene. Targeting of hPG80 with the humanized anti-hPG80 antibody has been shown:

1. a decrease in the self-renewal capacity of CSCs of various origins;

2. prolongation of in vitro and in vivo chemosensitivity and delayed relapse after treatment of T84 xenografted mouse cells;

3. decreased tumor burden and increased cell differentiation in the remaining tumors in transgenic mice developing Wnt-induced intestinal neoplasia.

This has recently been confirmed for ovarian, breast, esophageal, liver and gastric cancers making this therapeutic antibody a potential multi-cancer drug. [1]

Sensitization to radiotherapy

It has been shown that hPG80 is a radioresistant factor that can be targeted to sensitize rectum radiation-resistant cancers. [47] hPG80 decreased expression increases sensitivity to irradiation in colorectal cancer cell lines leading to increased radiation-induced DNA damage and apoptosis. In the same lineage, hPG80 targeting also results in radiation-induced survival pathways inhibition, Akt and ERK.

Related Research Articles

<span class="mw-page-title-main">Colorectal cancer</span> Cancer of the colon or rectum

Colorectal cancer (CRC), also known as bowel cancer, colon cancer, or rectal cancer, is the development of cancer from the colon or rectum. Signs and symptoms may include blood in the stool, a change in bowel movements, weight loss, and fatigue. Most colorectal cancers are due to old age and lifestyle factors, with only a small number of cases due to underlying genetic disorders. Risk factors include diet, obesity, smoking, and lack of physical activity. Dietary factors that increase the risk include red meat, processed meat, and alcohol. Another risk factor is inflammatory bowel disease, which includes Crohn's disease and ulcerative colitis. Some of the inherited genetic disorders that can cause colorectal cancer include familial adenomatous polyposis and hereditary non-polyposis colon cancer; however, these represent less than 5% of cases. It typically starts as a benign tumor, often in the form of a polyp, which over time becomes cancerous.

Autocrine signaling is a form of cell signaling in which a cell secretes a hormone or chemical messenger that binds to autocrine receptors on that same cell, leading to changes in the cell. This can be contrasted with paracrine signaling, intracrine signaling, or classical endocrine signaling.

<span class="mw-page-title-main">Gastrin</span> Mammalian protein found in Homo sapiens

Gastrin is a peptide hormone that stimulates secretion of gastric acid (HCl) by the parietal cells of the stomach and aids in gastric motility. It is released by G cells in the pyloric antrum of the stomach, duodenum, and the pancreas.

<span class="mw-page-title-main">Gastrin-releasing peptide</span>

Gastrin-releasing peptide, also known as GRP, is a neuropeptide, a regulatory molecule that has been implicated in a number of physiological and pathophysiological processes. Most notably, GRP stimulates the release of gastrin from the G cells of the stomach.

<span class="mw-page-title-main">Catenin</span>

Catenins are a family of proteins found in complexes with cadherin cell adhesion molecules of animal cells. The first two catenins that were identified became known as α-catenin and β-catenin. α-Catenin can bind to β-catenin and can also bind filamentous actin (F-actin). β-Catenin binds directly to the cytoplasmic tail of classical cadherins. Additional catenins such as γ-catenin and δ-catenin have been identified. The name "catenin" was originally selected because it was suspected that catenins might link cadherins to the cytoskeleton.

<span class="mw-page-title-main">Chondroblast</span> Mesenchymal progenitor cell that forms a chondrocyte

Chondroblasts, or perichondrial cells, is the name given to mesenchymal progenitor cells in situ which, from endochondral ossification, will form chondrocytes in the growing cartilage matrix. Another name for them is subchondral cortico-spongious progenitors. They have euchromatic nuclei and stain by basic dyes.

<span class="mw-page-title-main">Cripto</span> Protein-coding gene in the species Homo sapiens

Cripto is an EGF-CFC or epidermal growth factor-CFC, which is encoded by the Cryptic family 1 gene. Cryptic family protein 1B is a protein that in humans is encoded by the CFC1B gene. Cryptic family protein 1B acts as a receptor for the TGF beta signaling pathway. It has been associated with the translation of an extracellular protein for this pathway. The extracellular protein which Cripto encodes plays a crucial role in the development of left and right division of symmetry.

<span class="mw-page-title-main">TCF7L2</span> Protein-coding gene in humans

Transcription factor 7-like 2 , also known as TCF7L2 or TCF4, is a protein acting as a transcription factor that, in humans, is encoded by the TCF7L2 gene. The TCF7L2 gene is located on chromosome 10q25.2–q25.3, contains 19 exons. As a member of the TCF family, TCF7L2 can form a bipartite transcription factor and influence several biological pathways, including the Wnt signalling pathway.

<span class="mw-page-title-main">Gastrin-releasing peptide receptor</span> Protein-coding gene in the species Homo sapiens

The gastrin-releasing peptide receptor (GRPR), now properly known as BB2 is a G protein-coupled receptor whose endogenous ligand is gastrin releasing peptide. In humans it is highly expressed in the pancreas and is also expressed in the stomach, adrenal cortex and brain.

<span class="mw-page-title-main">LGR5</span>

Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) also known as G-protein coupled receptor 49 (GPR49) or G-protein coupled receptor 67 (GPR67) is a protein that in humans is encoded by the LGR5 gene. It is a member of GPCR class A receptor proteins. R-spondin proteins are the biological ligands of LGR5. LGR5 is expressed across a diverse range of tissue such as in the muscle, placenta, spinal cord and brain and particularly as a biomarker of adult stem cells in certain tissues.

<span class="mw-page-title-main">Protein Wnt-5a</span>

Protein Wnt-5a is a protein that in humans is encoded by the WNT5A gene.

<span class="mw-page-title-main">AXIN2</span> Protein-coding gene in the species Homo sapiens

Axin-2 also known as axin-like protein (Axil) or axis inhibition protein 2 (AXIN2) or conductin is a protein that in humans is encoded by the AXIN2 gene.

Mouse models of colorectal cancer and intestinal cancer are experimental systems in which mice are genetically manipulated, fed a modified diet, or challenged with chemicals to develop malignancies in the gastrointestinal tract. These models enable researchers to study the onset, progression of the disease, and understand in depth the molecular events that contribute to the development and spread of colorectal cancer. They also provide a valuable biological system, to simulate human physiological conditions, suitable for testing therapeutics.

<span class="mw-page-title-main">WNT2</span>

Wingless-type MMTV integration site family, member 2, also known as WNT2, is a human gene.

<span class="mw-page-title-main">TCF/LEF family</span> Group of genes

The TCF/LEF family is a group of genes that encode transcription factors which bind to DNA through a SOX-like high mobility group domain. They are involved in the Wnt signaling pathway, particularly during embryonic and stem-cell development, but also had been found to play a role in cancer and diabetes. TCF/LEF factors recruit the coactivator beta-catenin to enhancer elements of genes they target. They can also recruit members of the Groucho family of corepressors.

Kang-Yell Choi is a professor of biotechnology at Yonsei University, and has a joint appointment position as a CEO of CK Regeon Inc. in Seoul, Korea. He has been performing researches related to cellular signaling, especially for the Wnt/β-catenin pathway involving various pathophysiologies. Choi has been leading the Translational Research Center for Protein Function Control (TRCP), a Korean government supported drug development institute, as a director for 10 years. Choi has been carrying out R&D to develop agents controlling the Wnt/β-catenin signaling pathway. Choi's main interest is development of the agents to treat intractable diseases that suppress tissue regeneration system through overexpression of CXXC5 and subsequent suppression of the Wnt/β-catenin signaling.

<span class="mw-page-title-main">Apc, wnt signaling pathway regulator</span> Mammalian protein found in Homo sapiens

APC, WNT signaling pathway regulator is a protein that in humans is encoded by the APC gene.

<span class="mw-page-title-main">SOX7</span> Protein-coding gene in the species Homo sapiens

SRY-box 7 is a protein that in humans is encoded by the SOX7 gene.

Pro-Gastrin-Releasing-Peptide, also known as Pro-GRP, is a Gastrin-Releasing-Peptide (GRP) precursor, a neurotransmitter that belongs to the bombesine/neuromedin B family. GRP stimulates the secretion of gastrin in order to increase the acidity of the gastric acid. Pro-GRP is a peptide composed of 125 amino acids, expressed in the nervous system and digestive tract. It is also important to not confused with progastrin, consisting of 80 amino acids, precursor of gastrin in its intracellular version and oncogene in its extracellular version (hPG80).

Progastrin is an 80-amino acid intracellular protein and the precursor of gastrin, a gastrointestinal hormone produced by G cells in the gastric antrum. The main function of gastrin is to regulate acid secretion. During digestion, only gastrin is released into the bloodstream and stimulates the secretion of hydrochloric acid in the stomach as well as pancreatic digestive enzymes. In humans, progastrin is encoded by the GAST gene. Progastrin is expressed primarily in stomach tissue.

References

  1. 1 2 3 4 5 6 7 8 You, Benoit; Mercier, Frédéric; Assenat, Eric; Langlois-Jacques, Carole; Glehen, Olivier; Soulé, Julien; Payen, Léa; Kepenekian, Vahan; Dupuy, Marie; Belouin, Fanny; Morency, Eric (January 2020). "The oncogenic and druggable hPG80 (Progastrin) is overexpressed in multiple cancers and detected in the blood of patients". EBioMedicine. 51: 102574. doi:10.1016/j.ebiom.2019.11.035. ISSN   2352-3964. PMC   6938867 . PMID   31877416.
  2. 1 2 3 Hollande, F. (2003-04-01). "Adherens junctions and tight junctions are regulated via different pathways by progastrin in epithelial cells". Journal of Cell Science. 116 (7): 1187–1197. doi: 10.1242/jcs.00321 . PMID   12615962. S2CID   21931954.
  3. 1 2 Bardram, L.; Hilsted, L.; Rehfeld, J. F. (1990-01-01). "Progastrin expression in mammalian pancreas". Proceedings of the National Academy of Sciences. 87 (1): 298–302. Bibcode:1990PNAS...87..298B. doi: 10.1073/pnas.87.1.298 . ISSN   0027-8424. PMC   53250 . PMID   2296587.
  4. 1 2 3 van Solinge, W. W.; Odum, L.; Rehfeld, J. F. (1993-04-15). "Ovarian cancers express and process progastrin". Cancer Research. 53 (8): 1823–1828. ISSN   0008-5472. PMID   8467501.
  5. 1 2 3 Singh, P.; Xu, Z.; Dai, B.; Rajaraman, S.; Rubin, N.; Dhruva, B. (1994-03-01). "Incomplete processing of progastrin expressed by human colon cancer cells: role of noncarboxyamidated gastrins". American Journal of Physiology. Gastrointestinal and Liver Physiology. 266 (3): G459–G468. doi:10.1152/ajpgi.1994.266.3.g459. ISSN   0193-1857. PMID   8166285.
  6. 1 2 Singh, P.; Owlia, A.; Varro, A.; Dai, B.; Rajaraman, S.; Wood, T. (1996-09-15). "Gastrin gene expression is required for the proliferation and tumorigenicity of human colon cancer cells". Cancer Research. 56 (18): 4111–4115. ISSN   0008-5472. PMID   8797575.
  7. 1 2 3 Koh, Theodore J; Chen, Duan (September 2000). "Gastrin as a growth factor in the gastrointestinal tract". Regulatory Peptides. 93 (1–3): 37–44. doi:10.1016/s0167-0115(00)00176-2. ISSN   0167-0115. PMID   11033051. S2CID   26008211.
  8. 1 2 Siddheshwar, R K (2001-01-01). "Plasma levels of progastrin but not amidated gastrin or glycine extended gastrin are elevated in patients with colorectal carcinoma". Gut. 48 (1): 47–52. doi:10.1136/gut.48.1.47. ISSN   0017-5749. PMC   1728168 . PMID   11115822.
  9. 1 2 Brown, D.; Yallampalli, U.; Owlia, A.; Singh, P. (2003-01-01). "pp60c-Src Kinase Mediates Growth Effects of the Full-Length Precursor Progastrin1–80 Peptide on Rat Intestinal Epithelial Cells, in Vitro". Endocrinology. 144 (1): 201–211. doi: 10.1210/en.2002-220501 . ISSN   0013-7227. PMID   12488346.
  10. 1 2 Chakladar, Abhijit; Dubeykovskiy, Alexander; Wojtukiewicz, Lindsay J.; Pratap, Jitesh; Lei, Shi; Wang, Timothy C. (October 2005). "Synergistic activation of the murine gastrin promoter by oncogenic Ras and β-catenin involves SMAD recruitment". Biochemical and Biophysical Research Communications. 336 (1): 190–196. doi:10.1016/j.bbrc.2005.08.061. ISSN   0006-291X. PMID   16139800.
  11. 1 2 3 4 Pannequin, Julie; Delaunay, Nathalie; Buchert, Michael; Surrel, Fanny; Bourgaux, Jean–François; Ryan, Joanne; Boireau, Stéphanie; Coelho, Jessica; Pélegrin, André; Singh, Pomila; Shulkes, Arthur (November 2007). "β-Catenin/Tcf-4 Inhibition After Progastrin Targeting Reduces Growth and Drives Differentiation of Intestinal Tumors". Gastroenterology. 133 (5): 1554–1568. doi: 10.1053/j.gastro.2007.08.023 . ISSN   0016-5085. PMID   17920061.
  12. 1 2 Ramanathan, Vigneshwaran; Jin, Guangchun; Westphalen, Christoph Benedikt; Whelan, Ashley; Dubeykovskiy, Alexander; Takaishi, Shigeo; Wang, Timothy C. (2012-04-05). "P53 Gene Mutation Increases Progastrin Dependent Colonic Proliferation and Colon Cancer Formation in Mice". Cancer Investigation. 30 (4): 275–286. doi:10.3109/07357907.2012.657814. ISSN   0735-7907. PMC   3697930 . PMID   22480191.
  13. 1 2 Najib, S; Kowalski-Chauvel, A; Do, C; Roche, S; Cohen-Jonathan-Moyal, E; Seva, C (2014-08-11). "Progastrin a new pro-angiogenic factor in colorectal cancer". Oncogene. 34 (24): 3120–3130. doi: 10.1038/onc.2014.255 . ISSN   0950-9232. PMID   25109333. S2CID   20661274.
  14. 1 2 Giraud, J.; Failla, L. M.; Pascussi, J.-M.; Lagerqvist, E. L.; Ollier, J.; Finetti, P.; Bertucci, F.; Ya, C.; Gasmi, I.; Bourgaux, J.-F.; Prudhomme, M. (2016-05-04). "Autocrine Secretion of Progastrin Promotes the Survival and Self-Renewal of Colon Cancer Stem-like Cells". Cancer Research. 76 (12): 3618–3628. doi: 10.1158/0008-5472.can-15-1497 . ISSN   0008-5472. PMID   27197176.
  15. 1 2 3 4 5 Prieur, Alexandre; Cappellini, Monica; Habif, Guillaume; Lefranc, Marie-Paule; Mazard, Thibault; Morency, Eric; Pascussi, Jean-Marc; Flacelière, Maud; Cahuzac, Nathalie; Vire, Bérengère; Dubuc, Benjamin (2017-06-09). "Targeting the Wnt Pathway and Cancer Stem Cells with Anti-progastrin Humanized Antibodies as a Potential Treatment for K-RAS-Mutated Colorectal Cancer". Clinical Cancer Research. 23 (17): 5267–5280. doi: 10.1158/1078-0432.ccr-17-0533 . ISSN   1078-0432. PMID   28600477. S2CID   22085894.
  16. 1 2 Division of Gastroenterology, Department of Internal Medicine, VA Medical Center and University of Michigan, and the Michigan Gastrointestinal Peptide Research Center, Ann Arbor, MI 48109, USA Division of Gastroenterology, Department of Internal Medicine, VA Medical Center and University of Michigan, and the Michigan Gastrointestinal Peptide Research Center, Ann Arbor, MI 48109, USA Division of Gastroenterology, Department of Pediatrics, VA Medical Center and University of Michigan, and the Michigan Gastrointestinal Peptide Research Center, Ann Arbor, MI 48109, USA Division of Gastroenterology, Department of Internal Medicine, VA Medical Center and University of Michigan, and the Michigan Gastrointestinal Peptide Research Center, Ann Arbor, MI 48109, USA Kochman, Michael Lee DelValle, John Dickinson, Chris John Boland, C. Richard (2006-04-10). Post-translational processing of gastrin in neoplastic human colonic tissues. Elsevier. OCLC   894059948.{{cite book}}: CS1 maint: multiple names: authors list (link)
  17. Van Solinge, Wouter W.; Nielsen, Finn C.; Friis-Hansen, Lennart; Falkmer, Ursula G.; Rehfeld, Jens F. (April 1993). "Expression but incomplete maturation of progastrin in colorectal carcinomas". Gastroenterology. 104 (4): 1099–1107. doi: 10.1016/0016-5085(93)90279-l . ISSN   0016-5085. PMID   8462798.
  18. Ciccotosto, Giuseppe D.; McLeish, Andrew; Hardy, Kenneth J.; Shulkes, Arthur (October 1995). "Expression, processing, and secretion of gastrin in patients with colorectal carcinoma". Gastroenterology. 109 (4): 1142–1153. doi: 10.1016/0016-5085(95)90572-3 . ISSN   0016-5085. PMID   7557079.
  19. "Gastrin gene expression in human pancreatic and colon cancers". Gastroenterology. 108 (4): A1246. April 1995. doi:10.1016/0016-5085(95)29280-2. ISSN   0016-5085.
  20. Imdahl, A.; Eggstein, S.; Baldwin, G.S.; Farthmann, E.H. (April 1995). "Expression of gastrin, gastrin/CCK-B- and gastrin/CCK-C. Receptors in human colorectal carcinomas". Gastroenterology. 108 (4): A484. doi: 10.1016/0016-5085(95)26249-0 . ISSN   0016-5085.
  21. Nemeth, J; Taylor, B; Pauwels, S; Varro, A; Dockray, G J (1993-01-01). "Identification of progastrin derived peptides in colorectal carcinoma extracts". Gut. 34 (1): 90–95. doi:10.1136/gut.34.1.90. ISSN   0017-5749. PMC   1374107 . PMID   8432459.
  22. Cobb, Stephanie; Wood, Thomas; Ceci, Jeffrey; Varro, Andrea; Velasco, Marco; Singh, Pomila (2004-03-15). "Intestinal expression of mutant and wild-type progastrin significantly increases colon carcinogenesis in response to azoxymethane in transgenic mice". Cancer. 100 (6): 1311–1323. doi:10.1002/cncr.20094. ISSN   0008-543X. PMID   15022301. S2CID   2139822.
  23. Singh, Pomila; Velasco, Marco; Given, Randall; Wargovich, Michael; Varro, Andrea; Wang, Timothy C. (2000-03-01). "Mice overexpressing progastrin are predisposed for developing aberrant colonic crypt foci in response to AOM". American Journal of Physiology. Gastrointestinal and Liver Physiology. 278 (3): G390–G399. doi:10.1152/ajpgi.2000.278.3.g390. ISSN   0193-1857. PMID   10712258.
  24. Wu, Hai; Owlia, Azarmidokht; Singh, Pomila (December 2003). "Precursor peptide progastrin1-80 reduces apoptosis of intestinal epithelial cells and upregulates cytochrome c oxidase Vb levels and synthesis of ATP". American Journal of Physiology. Gastrointestinal and Liver Physiology. 285 (6): G1097–G1110. doi:10.1152/ajpgi.00216.2003. ISSN   0193-1857. PMID   12881229.
  25. 1 2 Westwood, David A; Patel, Oneel; Christophi, Christopher; Shulkes, Arthur; Baldwin, Graham S (2017-04-21). "Progastrin: a potential predictive marker of liver metastasis in colorectal cancer". International Journal of Colorectal Disease. 32 (7): 1061–1064. doi:10.1007/s00384-017-2822-8. ISSN   0179-1958. PMID   28432443. S2CID   4574556.
  26. Huels, D J; Sansom, O J (2015). "Stem vs non-stem cell origin of colorectal cancer". British Journal of Cancer. 113 (1): 1–5. doi:10.1038/bjc.2015.214. ISSN   0007-0920. PMC   4647531 . PMID   26110974.
  27. Hori, Hiroyuki Nakata, Hirohisa Iguchi, Genzo Yamada, Hajime Chihara, Kazuo Baba, Hisamitsu (2010-04-01). "Oncogenic ras induces gastrin/CCKB receptor gene expression in human colon cancer cell lines LoVo and Colo320HSR" (PDF). The Journal of Laboratory and Clinical Medicine. 神戸大学保健管理センター. 141 (5): 335–341. doi:10.1016/S0022-2143(03)00021-0. OCLC   997679353. PMID   12761477.{{cite journal}}: CS1 maint: multiple names: authors list (link)
  28. Cartwright, C. A.; Meisler, A. I.; Eckhart, W. (1990-01-01). "Activation of the pp60c-src protein kinase is an early event in colonic carcinogenesis". Proceedings of the National Academy of Sciences. 87 (2): 558–562. Bibcode:1990PNAS...87..558C. doi: 10.1073/pnas.87.2.558 . ISSN   0027-8424. PMC   53304 . PMID   2105487.
  29. Kondo, Jumpei; Powell, Anne E.; Coffey, Robert J. (2016). "466 Escape From BMP4-Mediated Growth Suppression Is an Early Event in Colonic Neoplasia". Gastroenterology. 150 (4): S102–S103. doi:10.1016/s0016-5085(16)30451-6. ISSN   0016-5085.
  30. 1 2 Ferrand, Audrey; Bertrand, Claudine; Portolan, Ghislaine; Cui, Guanglin; Carlson, Jane; Pradayrol, Lucien; Fourmy, Daniel; Dufresne, Marlene; Wang, Timothy C.; Seva, Catherine (2005-04-01). "Signaling Pathways Associated with Colonic Mucosa Hyperproliferation in Mice Overexpressing Gastrin Precursors". Cancer Research. 65 (7): 2770–2777. doi: 10.1158/0008-5472.can-04-0978 . ISSN   0008-5472. PMID   15805277.
  31. Rengifo-Cam, W.; Umar, S.; Sarkar, S.; Singh, P. (2007). "Antiapoptotic Effects of Progastrin on Pancreatic Cancer Cells Are Mediated by Sustained Activation of Nuclear Factor- B". Cancer Research. 67 (15): 7266–7274. doi: 10.1158/0008-5472.can-07-1206 . ISSN   0008-5472. PMID   17671195.
  32. Umar, S; Sarkar, S; Cowey, S; Singh, P (2008-06-02). "Activation of NF-κB is required for mediating proliferative and antiapoptotic effects of progastrin on proximal colonic crypts of mice, in vivo". Oncogene. 27 (42): 5599–5611. doi:10.1038/onc.2008.169. ISSN   0950-9232. PMC   2891442 . PMID   18521082.
  33. Pannequin, J.; Bonnans, C.; Delaunay, N.; Ryan, J.; Bourgaux, J.-F.; Joubert, D.; Hollande, F. (2009-07-21). "The Wnt Target Jagged-1 Mediates the Activation of Notch Signaling by Progastrin in Human Colorectal Cancer Cells". Cancer Research. 69 (15): 6065–6073. doi: 10.1158/0008-5472.can-08-2409 . ISSN   0008-5472. PMID   19622776.
  34. Singh, P.; Lu, X.; Cobb, S.; Miller, B. T.; Tarasova, N.; Varro, A.; Owlia, A. (2003-02-01). "Progastrin1–80 stimulates growth of intestinal epithelial cells in vitro via high-affinity binding sites". American Journal of Physiology. Gastrointestinal and Liver Physiology. 284 (2): G328–G339. doi:10.1152/ajpgi.00351.2002. ISSN   0193-1857. PMID   12388191.
  35. Dubeykovskiy, Alexander; Nguyen, Thomas; Dubeykovskaya, Zinaida; Lei, Shi; Wang, Timothy C. (2008). "Flow cytometric detection of progastrin interaction with gastrointestinal cells". Regulatory Peptides. 151 (1–3): 106–114. doi:10.1016/j.regpep.2008.07.001. ISSN   0167-0115. PMC   2630224 . PMID   18674570.
  36. Ottewell, P. D.; Varro, A.; Dockray, G. J.; Kirton, C. M.; Watson, A. J. M.; Wang, T. C.; Dimaline, R.; Pritchard, D. M. (2005). "COOH-terminal 26-amino acid residues of progastrin are sufficient for stimulation of mitosis in murine colonic epithelium in vivo". American Journal of Physiology. Gastrointestinal and Liver Physiology. 288 (3): G541–G549. doi:10.1152/ajpgi.00268.2004. ISSN   0193-1857. PMID   15486344.
  37. Singh, P; Wu, H; Clark, C; Owlia, A (2006-07-10). "Annexin II binds progastrin and gastrin-like peptides, and mediates growth factor effects of autocrine and exogenous gastrins on colon cancer and intestinal epithelial cells". Oncogene. 26 (3): 425–440. doi: 10.1038/sj.onc.1209798 . ISSN   0950-9232. PMID   16832341. S2CID   10850923.
  38. Sarkar, Shubhashish; Swiercz, Rafal; Kantara, Carla; Hajjar, Katherine A.; Singh, Pomila (2011). "Annexin A2 Mediates Up-regulation of NF-κB, β-catenin, and Stem Cell in Response to Progastrin in Mice and HEK-293 Cells". Gastroenterology. 140 (2): 583–595.e4. doi:10.1053/j.gastro.2010.08.054. ISSN   0016-5085. PMC   3031715 . PMID   20826156.
  39. Jin, Guangchun; Sakitani, Kosuke; Wang, Hongshan; Jin, Ying; Dubeykovskiy, Alexander; Worthley, Daniel L.; Tailor, Yagnesh; Wang, Timothy C. (2017-03-23). "The G-protein coupled receptor 56, expressed in colonic stem and cancer cells, binds progastrin to promote proliferation and carcinogenesis". Oncotarget. 8 (25): 40606–40619. doi: 10.18632/oncotarget.16506 . ISSN   1949-2553. PMC   5522213 . PMID   28380450. S2CID   4727270.
  40. Caplin, M.E.; Savage, K.; Khan, K.; Rode, J.; Brett, B.; Varro, A.; Michaeli, D.; Pounder, R.E.; Dhillon, A.P. (1998). "Expression and processing of gastrin in patients with hepatocellular carcinoma, fibrolamellar carcinoma and cholangiocarcinoma". Gastroenterology. 114: A1219. doi:10.1016/s0016-5085(98)84945-7. ISSN   0016-5085.
  41. Caplin, M.; Savage, K.; Khan, K.; Brett, B.; Rode, J.; Varro, A.; Dhillon, A. (2000-08-01). "Expression and processing of gastrin in pancreatic adenocarcinoma". British Journal of Surgery. 87 (8): 1035–1040. doi:10.1046/j.1365-2168.2000.01488.x. ISSN   0007-1323. PMID   10931047. S2CID   24292877.
  42. DuBois, Raymond N. (2009), "Cyclooxygenase-2 in Colorectal Cancer", Encyclopedia of Cancer, Berlin, Heidelberg: Springer Berlin Heidelberg, p. 799, doi:10.1007/978-3-540-47648-1_1436, ISBN   978-3-540-36847-2
  43. Paterson, Adrienne C; Macrae, Finlay A; Pizzey, Cathy; Baldwin, Graham S; Shulkes, Arthur (2014-02-19). "Circulating gastrin concentrations in patients at increased risk of developing colorectal carcinoma". Journal of Gastroenterology and Hepatology. 29 (3): 480–486. doi:10.1111/jgh.12417. ISSN   0815-9319. PMID   24716212. S2CID   25335053.
  44. Do, C.; Bertrand, C.; Palasse, J.; Delisle, M.-B.; Shulkes, A.; Cohen-Jonathan-Moyal, E.; Ferrand, A.; Seva, C. (2012-02-24). "A New Biomarker That Predicts Colonic Neoplasia Outcome in Patients with Hyperplastic Colonic Polyps". Cancer Prevention Research. 5 (4): 675–684. doi: 10.1158/1940-6207.capr-11-0408 . ISSN   1940-6207. PMID   22366915. S2CID   53420.
  45. Do, Catherine; Bertrand, Claudine; Palasse, Julien; Delisle, Marie-Bernadette; Cohen-Jonathan-Moyal, Elizabeth; Seva, Catherine (2013-11-08). "Activation of pro-oncogenic pathways in colorectal hyperplastic polyps". BMC Cancer. 13 (1): 531. doi:10.1186/1471-2407-13-531. ISSN   1471-2407. PMC   3829387 . PMID   24209454.
  46. Carr, Brian I; Pancoska, Petr; Branch, Robert A (2010-02-25). "Low alpha-fetoprotein hepatocellular carcinoma". Journal of Gastroenterology and Hepatology. 25 (9): 1543–1549. doi: 10.1111/j.1440-1746.2010.06303.x . ISSN   0815-9319. PMID   20796153.
  47. Kowalski-Chauvel, Aline; Gouaze-Andersson, Valerie; Vignolle-Vidoni, Alix; Delmas, Caroline; Toulas, Christine; Cohen-Jonathan-Moyal, Elizabeth; Seva, Catherine (2017-04-20). "Targeting progastrin enhances radiosensitization of colorectal cancer cells". Oncotarget. 8 (35): 58587–58600. doi:10.18632/oncotarget.17274. ISSN   1949-2553. PMC   5601677 . PMID   28938581.