Loading

Journal of Cancer Biology and Research

Aquaporins in Colorectal Cancer: Exploring Their Role in Tumorigenesis, Metastasis, and Drug Response

Review Article | Open Access | Volume 10 | Issue 2

  • 1. Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, USA
  • 2. Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, USA
  • 3. Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine (IBTO), Iran
  • 4. Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Iran
  • 5. Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Iran
  • 6. Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Iran
  • 7. Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Iran
  • 8. Department of Immunology, Department of Immunology School of Medicine, Iran University of Medical Sciences, Iran
  • 9. Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Iran
+ Show More - Show Less
Corresponding Authors
Elahe Safari, Department of Immunology, Iran University of Medical Sciences. Tehran. Iran
Abstract

Aquaporins (AQPs) are small, integral proteins facilitating water transport across plasma cell membranes in reaction to osmotic gradients. This family has thirteen unique members (AQP0-12), which can also transport glycerol, urea, gases, and other salute small molecules. AQPs play a crucial role in the regulation of different cellular processes, including metabolism, migration, immunity, barrier function and angiogenesis. These proteins are found to aberrantly overexpress in various cancers, including colorectal cancer (CRC). Increasing evidence has been explored AQPs as a potential diagnostic biomarkers and therapeutic targets in different cancers. However, there is no comprehensive review compiling the available information on the crucial role of AQPs in the context of colorectal cancer. This review highlights the significance of AQPs as the biomarker and regulator of tumor cells metabolism. In addition, the proliferation, angiogenesis, and metastasis of tumor cells related to AQPs expression and function are discussed. Furthermore, understanding the AQPs prominent role in chemotherapy resistance is of great importance as a clinical point of view.

Keywords

• Aquaporin

• Colorectal cancer

• Biomarkers

• Angiogenesis

• Metastasis

• Chemotherapy

CITATION

Mohammadabadi MA, Moazzeni A, Jafarzadeh L, Faraji F, et al. (2023) Aquaporins in Colorectal Cancer: Exploring Their Role in Tumorigen esis, Metastasis, and Drug Response. J Cancer Biol Res 10(2): 1140.

INTRODUCTION

Colorectal cancer (CRC) is known as the third most common cancer worldwide, and the second cause of death among cancer patients. In 2020, 10% of all cancer cases and 9.4% of cancer related deaths were due to colorectal cancer, meaning about 1.9 million new cases and 0.9 million human deaths [1-3]. In recent years, regarding the high prevalence and mortality rate of patients with colorectal cancer, the world has faced challenges of serious need for medical services. The prevalence of colorectal cancer has increased, especially in people less than 50 years of age, and about 2% of patients with colorectal cancer have metastases at the time of diagnosis, and more than 50% of patients with primary tumors will develop metastases in the future. Although the prognosis of patients has significantly improved in the last twenty years, however, metastatic colorectal cancer is still an incurable disease in most cases [4-6].

Currently, the treatment methods are complicated and associated with serious challenges; that even may lead to death. 

In metastatic types, the treatments are usually ineffective and are associated with the mortality and morbidity of the patients. Taken together, developing a new effective treatment for tumor cell growth and proliferation, invasion, and metastasis is of great importance [7].

Cell metabolism is dependent on different groups of molecules located in the cell membranes as transporters [8,9].

One group of molecules which are of special interest, are aquaporins (AQPs) [7]. Aquaporins are protein receptors, primarily known as water channels, with the ability to regulate the transport of water, glycerol, urea, and other small molecules. AQPs have been targeted in the treatment of different diseases such as diabetes insipidus and ADH syndrome. Recently, the role of these molecules in a much wider range of diseases has been identified, including cancer, heart failure, Sjögren’s disease, etc [10]. The structure and function, role in water physiology and, solutes movement make AQPs as suitable drug targets [8-11]. Besides maintaining the water balance, AQPs have special roles in cancer, including cell migration, proliferation, and adhesion and they are overexpressed on tumor cells and there is often a significant association between the expression of AQPs and tumor grade [9-12,13]. Consequently, AQPs can be targeted in cancer therapy by means of regulators.

Aquaporins structure

AQPs are small water-channel trans-membrane proteins that assist in the bi-directional transport of the water through the cell membrane. In addition, AQP can act as the transporter of other small neutral molecules such as glycerol and urea, or certain gases like carbon dioxide and ammonia as osmotic gradients in all cells [14].

The human AQPs consist of 13 mammalian proteins (AQ0 AQ12), which have unique cellular distribution. Interestingly, six of the thirteen AQP isoforms (AQP1, AQP2, AQP4, AQP5, AQP7 and, AQP10) have high-resolution structures [15-21].

The structure of AQP is consisting of six alpha helices domains which are stretch through the membrane with both the amino acid and carboxylic terminals on the cytoplasmic side. This structure is highly conserved in all living organisms [22]. The alpha helixes are connected by five loops from A to E. Loops B (Cytosolic) and E (Non-cytosolic) contain a highly conserved Asparagine-Proline Alanin (NPA) motif, which forms a barrel surrounding in the central pore-like region that contains additional protein density [14]. These motifs form a hourglass structure, making the water channel narrow in the middle (2.8 Å) and wider at each end.

The second section in non-cytosolic end of the pore is the “aromatic/Arginine (ar/R)” selectively filters. This filter consists of a cluster of amino acids, which enable the AQPs to selectively lead or block the transport of different molecules [22,23].Through the amino acid residues of the NPA and ar/R filters, alongside the other parts of the channel, AQP controls the substrate selection [14]. AQPs form a homo-tetramer structure in the cell membrane in which all four monomers act as a water channel alone. The helices of each AQP monomer which are located on the outer part of the tetramer, are hydrophobic, while the helices that are near the center of the tetramer are hydrophilic [24]. Functionally, based on permeability characteristics, AQPs members are divided into two groups [25]. 1: water selective or classic AQPs (AQP 1, 2, 4, 5, and 8) which facilitate water transport exclusively. 2: Aquaglyceroporins, which have the additional ability to direct small carbohydrates, especially glycerol (AQP 3, 7, 9, and 10). The two subgroups can be distinguished based on amino acid sequences. The aquaglyceroporins group includes glycerol facilitators (GlpFs), from glycerol permease facilitators. The “signature” sequence for aquaglyceroporins is the aspartic acid residue (D) in the NPA motif, which enlarges the pore to accept larger molecules [26]. The schematic illustration of AQP is depicted in Figure 1.

Schematic illustration of Aquaporin. A. Monomer topology of AQPs comprises of six tetramer helics 1-6, connected with loops A-E. Loops A, E and C are  extracellular, loops B and E are fold back to the cell membrane (with N-P-A codes that shows the NPA motifs and ar/R) and the intracellular N and C terminals B.  Monomer water channel formation by organization of helices and NPA motifs. C. Establishment of homotetramer with central pore, assembled by AQP monomers. D.  Crystal structure of the monomer and E. Homotetramer of AQPs viewed from the side of membrane. F. Homotetramer crystal structure from top view revealing four  water channel formed by each monomer and a central pore resulted of the homotetramer formation marked as X. Aquaporins: AQPs. Highl.y conserved Asparagine Proline-Alanin: NPA

Figure 1: Schematic illustration of Aquaporin. A. Monomer topology of AQPs comprises of six tetramer helics 1-6, connected with loops A-E. Loops A, E and C are extracellular, loops B and E are fold back to the cell membrane (with N-P-A codes that shows the NPA motifs and ar/R) and the intracellular N and C terminals B. Monomer water channel formation by organization of helices and NPA motifs. C. Establishment of homotetramer with central pore, assembled by AQP monomers. D. Crystal structure of the monomer and E. Homotetramer of AQPs viewed from the side of membrane. F. Homotetramer crystal structure from top view revealing four water channel formed by each monomer and a central pore resulted of the homotetramer formation marked as X. Aquaporins: AQPs. Highl.y conserved Asparagine Proline-Alanin: NPA

Aquaporins expression in normal and tumor tissues

AQP-0 is known as the major intrinsic protein (MIP) of the lens cells [27]. The mutations in MIP are predicted to disturb water flux across the lens cell membrane and causes cataracts [28]. Water transportation by AQP0 is at a slow rate; however, it has been demonstrated to play an important role in the cell-to-cell adhesion of the lens fiber. AQP1 as the most studied aquaporin is the first with a high-resolution structure. AQP1 is expressed in various tissues e.g., lungs, kidneys and, red blood cells. Mutations in AQP1 are shown to be related to urinary concentration deficiency or failure in peritoneal dialysis [29-30]. AQP1 is also up-regulated in different cancers, including multiple myeloma, lung, and colon and brain tumors, mammary carcinoma, choroid plexus tumors and hemangioblastoma [31-39].

AQP2 was found in renal collecting duct, and called as the water channel of the collecting duct [14]. AQP2 is shown as a diagnostic marker for pheochromocytoma and/or paraganglioma and is expressed in glioma cell lines [40-41].

AQP3 is expressed in the skin and provides cells glycerol. It is shown that the deletion of AQP has resulted in impairs of elasticity, skin hydration, barrier recovery and wound healing in which the administration of glycerol has reversed it [42]. It is found to abnormally express in various cancers including breast cancer, gastric cancer, colorectal cancer, and melanoma and plays a pivotal role in cancer metastasis [43]. AQP4 is the main water channel which express in the central nervous system [44]. In edematous astrocytomas and metastatic tumors, AQP4 is demonstrated to be up-regulated [45]. AQP5 controls the water flux throughout several body systems including digestive, respiratory, renal, integumentary, and reproductive systems and also in sense organs [13]. The AQP5 expression is associated with different cancers including lung, breast, colorectal and ovarian cancers [46-49]. AQP6 is found as anion permeation with marked specificity for nitrate which is shown to express in epithelial ovarian tumors [50-51]. AQP7 is localized to a wide range of tissues [52] and is shown to regulate the response to cellular stress in breast cancer [53]. AQP8 expression in various cancers is associated with clinical significance [54-56]. AQP9 as the glycerol transporter is strongly expressed in human astrocytomas [12]. and its expression is correlated with tumor grade in epithelial ovarian cancer (57). AQP10-12 are shown to express in both the normal and tumor tissues, however, much more attention needs to clarify their role in the context of different diseases [58,59].

Role of aquaporins in cancer proliferation, angiogenesis, and metastasis of CRC

Currently, it has been revealed that the expression of AQPs is increased in cancerous cells of different origins, particularly in invasive malignancies, such as CRC [60]. In addition, it has been shown that AQPs play an important role in tumor cell proliferation, angiogenesis, and migration [39-60,61]. AQP1 is demonstrated to express in human HT20 colon cancer cells and play role in tumor cell migration. The elevated expression of this AQP in HT20 cells leads to increase plasma membrane water permeability and cell migration in both invasive trans well and wound healing experiments, while, low expression level has reduced the plasma membrane water permeability and cell migration potency. Actin protein re-localization and RhoA and Rac activation were induced by AQP1 expression. Furthermore, following tail vein injection, AQP1 is shown to enhance the extravasation of HT20 cells in the lung of nude mice. In conclusion, the plasma membrane water permeability mediated by AQP, play crucial role in CRC metastasis and invasion [62]

. In HT29 colon cancer cells with increased expression of AQP1, cell migration is significantly inhibited, while no effect was detected on the migration rate of HCT-116 cells with low expression of AQP1. In addition, HT29 cells treated with AQP1 inhibitor showed a significantly reduced rate of invasion. Furthermore, in angiogenesis assay, this treatment resulted in complete inhibition of endothelial tube formation [63].

Tissue microarray (TMA) analysis is used to evaluate the association of clinic-pathological findings with AQP1 expression. Analysis of 120 stage II and III colon cancer patients showed 35.8% AQP1-positive rate. There was an association between the expression of AQP1 and lymph node metastasis and lymphovascular and vascular invasion [64]. Moon et al., has examined the expression of AQPs 1, 3, and 5 in seven colon and colorectal cancer cell lines using transcriptase–polymerase chain reaction analysis and in situ hybridization experiments. They found that in colorectal carcinogenesis, induction of AQPs 1 and 5 expressions was initiated in the early stage of the disease and maintained during the late stages of its development. Expression of these two AQPs was also maintained in metastasis to the liver. These results showed that AQPs 1 and 5 expressions are involved in the early stage of colorectal cancer development [65]. The Cancer Genome Atlas analysis has revealed that AQP1 expression was significantly decreased in CRC. However, following disease progression, the expression increased and was significantly elevated in stage IV compared to stages I and II, and also in patients with lymph node metastasis compared to those without [66]. The clinical significance of AQPs 1, 3, and 5 expressions in patients with CRC showed a significant correlation between these AQPs and lymph node metastasis in surgically resected CRC patients [49]. Li et al., examined whether AQP3 could increase the migration of human colorectal carcinoma cells or not. Additionally, its role in the prognosis of the disease was evaluated. Results revealed that AQP3 expression is up-regulated in CRC compared to the normal ones. In addition, there was a correlation between the levels of AQP3 expression and differentiation, lymph node, and distant metastasis of CRC. Human epidermal growth factor (hEGF), enhanced the expression of AQP3, followed by an increased migration rate of human colorectal cancer cells. Moreover, blocking of AQP3 and EGFR pathways inhibited the migration of CRC cells [67].

Different studies have shown that AQP5 has an important role in the proliferation and metastasis of CRC [65-68,69]. Clinical results indicate that enhanced expression of AQP5 in CRC is a predictor of tumor metastasis and is associated with poor prognosis [49-56,70,71]. CRC, the AQP5 overexpression is shown to be associated with increased circulating tumor cells and metastasis rate enhancement [71]. Moreover, it is reported that there is a close correlation between AQP5 expression and colorectal cancer cell differentiation, TNM stage, and metastasis. Over-expression of this AQP is associated with advanced TNM stage, lymph node metastasis, and poor prognosis [71].

The expression of AQP5 in 62.8% (59/94) of resected CRC tissue specimens and its relationship with liver metastasis is reported [69]. It is revealed that the increased expression of this AQP promotes the proliferation of CRC cells by induction of the Ras-MAPK pathway, whereas deletion of AQP5 leads to inhibition of cancer cell growth [69]. Deletion of AQP5 results in the inhibition of migration and invasion of CRC cells and modulates epithelial–mesenchymal transition (EMT) of CRC by suppressing the Wnt/b-catenin pathway [72]. Furthermore, the effects of AQP5 expression silencing in CRC cell lines resulted in decreased expression and secretion of vascular endothelial growth factor (VEGF)-A in these cells. Also, proliferation and angiogenesis of xenograft tumors were suppressed by inhibition of endogenous AQP5 expression in HT29 cells [73]. Examining the expressions of AQP5 mRNA and protein in CRC tissue and the effects of silencing protein expression of this AQP in the HT29 cell line showed that suppression of AQP5 can lead to inhibition of HT29 cell growth [74]. In another study, the knockdown of AQP5 decreased the proliferation of CRC [68]. These findings suggest that AQP5 may be involved in the detachment of tumor cells from the primary tumor to metastasize [75].

Investigating AQP8 expression in CRC revealed that the increased expression of this AQP significantly reduced the proliferation, aggressiveness, and colony formation of the SW480 and HT-29 cells following inactivation of PI3K/AKT signaling and inhibition of PCDH7 expression. In addition, studies on nude mice xenograft and metastasis models showed an important role for AQP8 in CRC cell proliferation and metastasis [76]. AQP8 is significantly expressed by para-neoplastic normal tissues and rarely by colorectal carcinoma cells; demonstrating downregulation of AQP8 in CRC tissues [56]. Recently, AQP9 is shown to highly express in metastatic CRC and causes Wnt/β catenin pathway activation through Dishevelled 2 (DVL2). This pathway eventually leads to EMT activation and invasion and migration of CRC cells [77]. Figure 2 is representative of AQPs role in tumor progression.

Aquaporins role in proliferation, angiogenesis, and metastasis of colorectal cancer.  Aquaporins: AQP. human Epidermal Growth Factor: hEGF. Vascular Endothelial Growth Factor: VEGF.Ras/Mitogen Activated Protein Kinase:Ras/MAPK. Epithelial Mmesenchymal Transition: EMT.  Dishevelled 2: DVL2.

Figure 2: Aquaporins role in proliferation, angiogenesis, and metastasis of colorectal cancer.

Aquaporins: AQP. human Epidermal Growth Factor: hEGF. Vascular Endothelial Growth Factor: VEGF.Ras/Mitogen Activated Protein Kinase:Ras/MAPK. Epithelial Mmesenchymal Transition: EMT. Dishevelled 2: DVL2.

Aquaporins in cellular metabolism

It is well-accepted that the growth, development, invasion, and metastasis of cancerous cells are closely related to the tumor microenvironment and cell metabolism. In addition, it has been shown that water molecules play crucial role in development of tumor cells and open insights into new strategies for cancer treatment. Increased water transport is required to meet the high metabolic demands of rapidly proliferating tumor cells. AQPs are involved in several physiological and metabolic mechanisms related to water and glycerol transport, and lipid metabolism. Additionally, as mentioned before, in tumors, the level of AQPs expression is high and abnormal. Therefore, AQPs via transfer of water and other molecules or small solutes, have a crucial role in different cellular functions such as regulation of the size and shape, energy metabolism, migration, adhesion, proliferation, and differentiation. Any disorders lead to edema, proliferation and migration of tumor cells as well as tumor angiogenesis and metastasis. Therefore, identifying the role of AQP as a transporter in homeostasis and cellular environments of tumors seems to be necessary, especially as a clinical point of view [78-81].

The pathological conditions of many diseases are associated with altered aquaporin function and abnormal AQP expression [82]. Aquaglyceroporins are involved in metabolic disorders such as obesity, diabetes and, etc. Disturbance in the ability of aquaglyceroporins for glycerol penetration, as well as their abnormal expression in certain tissues, is demonstrated to result in fat metabolism, cell proliferation, and epidermal water retention [83-84]. Orthodox aquaporins and aquaglyceroporin isoforms are associated with various cancers, often revealing a strong correlation between these AQPs expression levels and tumor grade [12-85]. In addition, the role of aquaglycerolporins in glycerol transport and its importance in cell proliferation and fat metabolism, and cancer progression make aquaglycerolporins a potential therapeutic target [86]. Aquaglycerolipins (AQPs 3, 7, 9, 10, and 11) affect the metabolic pathways and are highly expressed in tissues involved in glycerol metabolism, such as fat cells and the liver [87].

Collectively, increased aquaglyceroporin expression in tumors suggests that glycerol contributes to tumor growth and proliferation in two ways: first as a primary material in the synthesis of the phospholipids, and/or second as a mediator or regulator of ATP production [11]. For rapid cell proliferation in cancer, both pathways are necessary [86]. AQP3 is the most studied one among the four human aquaglyceroporin isoforms in cancers, which is expressed in different epithelial cells [88 89]. Inhibiting the AQP3 expression leads to a decrease in cell proliferation of different cell lines and silencing the AQP3 reduces some lipid synthases in gastric cancer cells. Therefore, the disturbance in lipid synthesis due to the reduction of AQP3 is not only due to the reduction of glycerol absorption but also related to the inhibition of the lipid synthesis system. The PI3K (phosphatidylinositol-4,5-bisphosphate 3-kinase)/Akt (protein kinase B) signaling pathway was also inhibited following destruction of AQP3. This pathway is involved in impaired lipid and ATP production [90].

Since tumor cells divide more rapidly, they need more lipids for membrane synthesis and more energy to continue their aggressive proliferation and malignancy (91, 92). Phospholipids are necessary for plasma membrane formation, but they can also be catabolized by β-oxidation to produce ATP. Triglycerides (TAG) produced by glycerol metabolization are vital for cell survival and proliferation. By lipolytic processes, TAG can convert into free fatty acid (FFA) in tumor cells. Fatty acid oxidation (FAO) for ATP production, promotes cancer development. Therefore, inhibiting or down-regulating aquaglyceroporins may lead to defects in supplying materials and energy for cancer cell growth following the inhibition of lipid synthesis [86-93]. Another hypothesis for the function of AQP3 in tumor cell development is overexpression of AQP3 in which causes greater permeability to glycerol and more ATP [94]. which is necessary for biosynthesis and tumor genesis. The positive correlation of glycerol level with the amount of ATP is evidence of this hypothesis [95]. Glycerol could be considered as an important regulator of ATP in cells which give the reason for the AQP3 overexpression in several tumors [86].Tumors use an un-regulated metabolism for cell proliferation and survival due to high nutrient requirements as sustaining a balanced redox state. Glucose and glutamine are usually important sources of energy in cancer. Warburg, in the 20s, proposed the theory that disruption of mitochondrial respiration causes tumor formation and increased glucose consumption, which resulted in high production of lactate, i.e., aerobic fermentation known as the Warburg effect [96]. The primary material for tumor cell proliferation can be prepared following the catabolism of glucose and glutamine [97]. Glucose induces carbon skeletons oxidation which causes electrons absorption by cells in the form of reduced nicotinamide. The malate-aspartate shuttle helps the adenine dinucleotide (NADH) to enter the mitochondrial electron transport chain. This process helps in cellular respiration and ATP production. Proliferating tumor cells promote glucose metabolism and convert excess pyruvate to lactate. This process, leads to maintaining the cytoplasmic level of the NAD+/NADH ratio [97-98]. For ATP production, glycerol can be used as an intermediate for pyruvate via the glycerol 3-phosphate shuttle (G-3-P shuttle) which results in tumor cell aquaglyceroporins overexpression. It should be noted that the cancer metabolism is constantly reprogrammed and optimized for available nutrient usage. Generally, glycerol as an intermediate of phospholipids can contribute to tumor cell proliferation, and its mutual effect on ATP production should be considered [86]. AQP9 is highly expressed in human glioblastoma, which reveals its role in the excretion of glycerol and lactate, indicating the involvement of lactic acidosis in glioma metabolism of energy. This confirms the important function of AQPs in glycerol absorption, as well as the regulation of its efflux [99]. It is shown that AQP9 is expressed in the basolateral membrane of benign and borderline tumor cells in ovarian cancer, whereas it is expressed widely in the membrane of malignant cells [57]. Additionally, it is demonstrated that a malignant ovary has more expression than a borderline which is more than benign/normal tissue. Also, AQP9 was overexpressed in mucinous tumors more than in serous ovarian tumors, and high-grade tumors revealed more expression than low grades [57-100], indicating that high expression of AQP9 could be correlated with poor prognosis. Consequently, this evidence reveals the importance of aquaglyceroporins and glycerol function in cancer development.

Although, AQP9 has lower expression in hepatocellular carcinoma [101], but its high expression can inhibit cell invasion and cancer development. Therefore, different roles and function of aquaglyceroporins may be seen in various cancers [86]. The tumor requirement of glycerol is associated to the type and stage of the cancer. In melanoma, the malignant phenotype is correlated with low AQP3 expression compared to normal skin and benign nevi. This shows that the transition from tumor proliferation to migration reduces the glycerol requirement [86]. Glycerol consumption increases in several non-metastatic cancer cell lines in contrast to metastatic ones [86]. AQP-5 can change the microenvironment and metabolism of tumor cells to increase the volume of liquid secretion from different tissues and result in the decrease in protein and salt concentration [84], and the proliferation of tumor cells. Collectively, it should be mentioned that glycerol requirement is different and depend on various tissue origins, malignancies, grades, and metastatic characteristics, which challenge aquaporin’s function and role. Additionally, due to the various role of AQPs, cancer requirement to control glycerol amount may lead to low expression of special isoforms [86].

Aquaporin role in CRC chemotherapy resistance

One main approach in CRC therapy is chemotherapy, especially for invasive types; however, the efficacy is affected by several variables. Cancer cells resistance to chemotherapy is one of the major reasons for its ineffectiveness. Chemotherapy resistance can be induced by several cellular factors and complex pathways [102-104]. Therefore, identifying the genes and mechanisms related to the development of multidrug resistance (MDR) is very important and can contribute to the treatment of CRC. Multidrug resistance can be classified into two groups. First, the MDR mechanism which consists of energy-dependent drug pumps. This MDR mechanism is mediated by special membrane glycoproteins, including P-glycoprotein 1 (P-gp), multidrug resistance-associated protein (MRP), and lung resistance-related protein (LRP). The second MDR mechanism consist of enzymes, such as DNA topoisomerase II (TOPO II), thymidylate synthase (TS), and protein kinase C [105-106]. AQPs are suggested to play a crucial role in MDR. Following assessment of the AQP5 expression levels and multidrug resistance genes, such as P-glycoprotein (P-gp), topoisomerase (TOPO), glutathione S-transferase-π (GST-π) and thymidylate synthase, it has been revealed that, the AQP5 can acts as a crucial regulator in multidrug resistance development in CRC. In addition, RNA interference technology is used to silence the AQP5 expression in differentiated human colon adenocarcinoma cell lines. Results revealed the crucial role of AQP5 in MDR development. These findings can be the basis for a potential effect of pharmacological AQP5 inactivation on chemotherapy sensitivity for colon cancer treatment [74].The sensitivity of HT-29 cells to the chemotherapeutic drugs including, cisplatin (DDP) and 5-fluorouracil (5-FU) is demonstrated to be increased following AQP5 silencing using siRNA. In addition, the higher levels of AQP5 expression are demonstrated to be associated with resistance to the imatinib mesylate, as a tyrosine kinase inhibitor, which is used in treatment of Chronic Myeloid Leukemia (CML) in the chronic phase. Blocking of AQP5, also reduced the cell proliferation rate in CML cells [107].MAPK [108-111], and PI3K/ Akt [112]. signaling pathways are involved in MDR. MAPK, serine/threonine protein kinase in eukaryotic cells, sends stimulating signals to the cells and induces reactions after activation. MAPK signaling pathways are composed of ERK, JNK/ stress-activated protein kinase (SPAK), and p38 MAPK [113]. PI3K/Akt is a crucial pathway for cell growth regulation [113]. AQP5 functions in the CRC MDR via controlling the resistance proteins. To determine the special signaling pathways that cause MDR by AQP5 in CRC, MAPK phosphorylation levels were measured in transfected HT-29 cells. It was shown that silencing of AQP5 expression reduces p38 MAPK phosphorylation and activation, indicating that, MAPK phosphorylation serves as a signaling pathway leading to MDR by AQP5 in CRC. Therefore, AQP5 inactivation may be suitable for drug-resistance treatment in CRC [74]. Recently, it has been shown that several AQPs induce diffusion of H2O2 through lipid cell membranes; including peroxyporins (AQP1, AQP3, AQP5, AQP8, AQP9 and, AQP11) [114-116]. The correlation between peroxyporins and cancer development has been shown in various studies [117]. Oxidative stress can cause genetic instability, and by changing cellular processes, CRC will eventually occur. On the other hand, adaptive oxidative defense induces therapeutic resistance, which causes failure in cancer treatment. Peroxyporins are known as aquaporin membrane channels that facilitate membrane permeation of H2O2 and are crucial for regulating cell proliferation and antioxidant defense. In a study, several colon cancer cell lines were treated with H2O2 and the sensitivity of cells to H2O2, cellular antioxidant status, intracellular ROS accumulation and the expression of AQP1, AQP3, and AQP5 peroxyporins were investigated. Results showed that HT-29 cells were the most resistant cell line and showed highly expression of peroxyporins and low levels of intracellular ROS, but in GSH levels, catalase activity, NF2, and PPARγ levels did not show any differences. This study shows that resistance to oxidative stress can be induced by different strategies except the antioxidant defense system. Peroxyporin can change the cellular antioxidant defense system and causes resistance to oxidative stress. Since regulation of H2O2 permeation can contribute to the tumor-resistant phenotype, controlling the intracellular ROS via regulating peroxyporin expression may be an important approach against MDR in cancer [118]. Little is known about the mechanism of AQPs as regulators of apoptotic genes. In addition, it is ambiguous whether the AQPs is located in the mitochondrial membrane and is involved in the apoptosis process by modulating water movement [119-120]. Several members of the AQP family are shown to aberrantly express in melanoma. AQP3 and AQP9 prevent the therapeutic effect of arsenite in melanoma by increasing the expression of anti-apoptotic genes including, Bcl-2 and XIAP while decreasing the expression of pro-apoptotic genes including, P53 and Bax; implying their anti-apoptotic role [106]. The presence of AQP3 in the nucleus confirms its association with apoptotic genes, which is probably different from their basic role as water transporters [106]. Although Cisplatin (cDDP) is in first line of chemotherapy drug for the treatment of gastric cancer (GC), but it faces different challenges of resistance. There are different and complex mechanisms for cDDP resistance. AQP3 is overexpressed in various cancers such as GC and is thought to have a function in GC development and progression. It is shown that AQ3 mediates cisplatin resistance in gastric cancer via autophagy. Therefore, AQP3-targeted therapies can be a suitable approach for GC treatment [121]. Autophagy is a special cell strategy with catabolic functions. This highly regulated mechanism induces the degradation of different intracellular components such as toxic aggregates, misfolded proteins, and damaged organelles, resulting in oxidative stress decrease and cell damage prevention [122]. Moreover, autophagy is induced in various situations, such as hypoxia, metabolic stress, nutrient deprivation, cancer treatments, and radiation therapy, and also for cellular adaptation and growth [123]. Autophagy may have a tumor suppressor role in normal cells, and dysregulation of this process may induce tumor development. In cancers, autophagy may lead to tumor progression due to its role in cellular adaption and survival. Autophagy is shown to induce chemoresistance in different cancers. In GC, autophagy which induces via AQP3 can lead to cDDP resistance [121-124]. Approximately, half of the breast cancer patients do not respond to initial chemotherapy treatments, which could be due to drug resistance. The effects of AQP-5 on drug resistance in breast cancer cells revealed enhanced drug sensitivity [125]. Additionally, more studies have confirmed the association of AQPs with sensitivity to chemotherapy drugs and better prognosis. The expression of AQP1 can be a predictive biomarker of response to chemotherapy in patients with CRC, and chemotherapy is ineffective in CRC patients without AQP1 expression. AQP1 has also been shown to be involved in the development, progression, and metastasis of various types of cancers, including CRC, which is associated with invasion and metastasis. However, its expression may be related to sensitivity to chemotherapy [126]. The role of AQP1 as a predictor of response to chemotherapy in breast cancer treatment is assessed. Results revealed that breast cancer patients, who gave anthracycline treatment in accordance with high expression of AQP1, had better responses in comparison to those with low expression of AQP1. Moreover, miR-320a-3p is shown to inhibit the expression of AQP1 and reduce the sensitivity to anthracycline drugs [127].

Aquaporins as biomarker in CRC

Biomarkers play significant roles in cancer diagnosis, prognosis, prediction, pharmacodynamics, and recurrence. AQPs have been identified in different cancers as biomarker with various roles [69,128-131]. In CRC, different groups of all thirteen AQPs isoforms are shown to be expressed [65,132-134]. It is proposed that, AQP5 is a prognostic biomarker for CRC regarding its high expression in 40 colorectal cancer tissues; related to tumor nodes-metastasis (TNM), stage, and differentiation in comparison to para-neoplastic tissues [56]. The AQP5 overexpression in specimens, from 45 colorectal cancer patients, can act as a biomarker related to TNM stage, circulating tumor cells (CTCs), metastasis, aggressiveness and predictive marker of prognosis [71-135]. High expression of AQP1 in resected cancer specimens of CRC patients is seen. Evaluation of 268 CRC specimens with different stages (0-IV) has revealed the high expression of AQP1 in about 41% of patients. Moreover, AQP1 expression also is significantly accompanied by multiple invasive features including; vessel and lymphatic invasion, lymph node metastasis, greater invasive depth, and also the differentiation level. However, the expression of AQP1 is not associated with hepatic metastasis [126]. AQP1 is suggested as a predictive biomarker to respond to 5-fluorouracil-based adjuvant chemotherapy in CRC patients with stage II and III cancer [126]. Therefore, adjuvant chemotherapy in patients with CRC is recommended based on the expression pattern of AQP1, suggesting it as an ideal target for patients with CRC in stages II and III of the disease [136]. The association of AQP1 expression with a lower 5-year survival rate and higher invasiveness in 120 CRC patients in stages II and III is indicative of AQP1 as a poor prognostic biomarker [64]. Lower expression of AQP9 has been proposed as a predictive biomarker in response to adjuvant chemotherapy in patients with CRC in stage III [137]. Furthermore, AQP 1 and 5 are found in the early stage of CRC development; suggesting their role as progression biomarkers [65].

Therapeutic Potential of Aquaporin in CRC

AQPs are shown to play a role in tumor invasiveness. Table 1 demonstrates the therapeutic potential of AQPs inhibition. Given that overexpression of AQP1 is related to proliferation, angiogenesis, and lymph node metastasis of cancer cells in CRC, it could be considered as an appropriate therapeutic target. Pharmacologically and/or genetically inhibition of AQP1 could suppress the invasion, proliferation, and growth of other types of cancer cells such as breast, prostate, thyroid, bladder, and ovarian cancers [138-141]. AQP1 has been targeted in various cancers by miR-223 [139], miR-133a-3p [142], Rg3 [140], AqB007, AqB011 [143], AqB013 [63]. and, J82 [144]. AqB013 is one example of drug used to evaluate the effect of AQP1 blockade in the treatment of CRC. Treatment of HT29 colon cancer cells, expressing a high level of AQP1 with AqB013 have demonstrated significant suppression in migration, invasion, and complete angiogenesis blockade in a dose-dependent manner, while, it has no effect on migration of HCT-116 cells with low AQP1 expression [63]. Following the assessment of the effect of Acetazolamide in CRC xenograft of nude mouse, it is found that this drug is able to reduce tumor growth by decreasing the expression of AQP1 at the level of RNA and protein. It is noteworthy that, inhibiting AQP1 expression was related to inhibition of VEGF expression as a known angiogenic factor in cancer [145]. The effect of aquaporin ligand; bumetanide derivatives including AqB001, AqB006, AqB007, and AqB011 were evaluated following AQP1 obstruction in the human HT29 cell line [143]. AqB011 and AqB007 by interacting with D loop domain in the AQP1 structure revealed the greatest inhibition of ion conductance of AQP1 which led to the reduction of migration and metastasis of CRC cell lines [143]. It has been revealed that AqB011 and bacopaside II by inhibition of ion conductance and water channel activity of AQP1 (respectively) lead to augment the inhibition of colon cancer cell migration [146]. Targeting microRNAs tend to be another method of inhibiting protein expressions in different cancers [147-149]. It has been stated that AQP1 overexpresses in colon cancer cells [142] can serve as one of the miR-133-39p targets [150]. miR-133a-3p overexpression significantly suppresses AQP1 expression which lead to a decrease in cell proliferation, migration, and invasion in CRC cells [142]. Inhibition of AQP3 using nano-formulations of a potent copper-based aquaporin inhibitor have shown to increase the cytotoxic effect on tumor cells of C26 murine cells [151]. The blockade of AQP3 expression using CuSO4 is resulted in enhanced migration of HCT116 cells. Additionally, the hEGF induction of AQP3 overexpression is demonstrated to be suppressed using PI3K/AKT inhibitor, LY294002. However, U0126 as the inhibitor of ERK caused a minor effect on the hEGF induced AQP3 up-regulation [67]. Silencing the AQP5 in CRC cell lines is shown to be related to impaired migration and invasion of colorectal cancer cells. In addition, the epithelial–mesenchymal transition (EMT) markers were altered as the expression of E-cadherin, Tissue Inhibitor of Metalloproteinases (TIMP)-1 and TIMP-2 were increased, while the Vimentin, N-cadherin, Plasminogen Activator, Urokinase (uPA) and Snail were down regulated. The Wnt/β-catenin pathway was markedly changed toward reduction, indicating the effect of AQP5 expression on invasiveness and metastasis of colon cancer [72]. Cairicoside E, a natural resin glycoside compound is revealed to suppress the AQP5 expression which results in EMT inhibition and a further reduction in p-Smad2/3 induced by TGF-β1. It is well confirmed that the effect of Cairicoside E is on AQP5 expression in which the CE had no significant effect on EMT markers and p-Smad2/3 induced by TGF-β1 in the condition of AQP5 silencing, separately [152]. Silencing the AQP5 using a specific short hairpin RNA construct could increase the chemosensitivity of CRC cells to 5-fluorouracil, mediating the apoptosis and suppression of tumor growth. The 5-fluorouracil (5-FU) chemosensitivity is mediated by the Wnt–β-catenin pathway suggesting AQP5 as a useful therapeutic target for CRC [153]. Berberine as a natural isoquinoline alkaloid is shown to have anti-cancer properties. Findings indicate that, the administration of this molecule lead to inhibition of the migration, invasion and the growth of CRC cells via down-regulation of AQP 1, 3 and, 5 expressions. This process is mediated following up-regulation of PTEN which inhibits the PI3K/AKT pathway, both at the gene and protein levels [35].

CONCLUSION

Aquaporins tend to be considered crucial in the development, angiogenesis, and invasiveness of colorectal cancer as one of the major concerns of the world. Among the thirteen isoforms, AQP1, 3 and, 5 seem to be related to poor prognosis of CRC, and their aberrant expression is associated with high invasiveness and increased tumor cell proliferation, angiogenesis, and metastasis. They also can serve as the reason for chemotherapy ineffectiveness. In addition, by regulating the cellular metabolism, the AQP3 and 9 play a crucial role in tumor genesis. Taken together, a better understanding of aquaporin role in colorectal carcinogenesis leads to better identification of novel AQP inhibitors to target specific AQPs associated with CRC cancers.

REFERENCES

1. Weng J, Li S, Zhu Z, Liu Q, Zhang R, Yang Y, et al. Exploring immunotherapy in colorectal cancer. J Hematol Oncol. 2022; 15: 1-28.

2. Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, et al. Colorectal cancer statistics. CA Cancer J Clin. 2020; 70: 145-64.

3. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022; 72: 7-33.

4. Ciardiello F, Ciardiello D, Martini G, Napolitano S, Tabernero J, Cervantes A. Clinical management of metastatic colorectal cancer in the era of precision medicine. CA Cancer J Clin. 2022; 72: 372-401.

5. Biller LH, Schrag D. Diagnosis and treatment of metastatic colorectal cancer: a review. JAMA. 2021; 325: 669-85.

6. Kocarnik JM, Compton K, Dean FE, Fu W, Gaw BL, Harvey JD, et al. Cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life years for 29 cancer groups from 2010 to 2019: a systematic analysis for the Global Burden of Disease Study 2019. JAMA Oncol. 2022; 8: 420-44.

7. De Ieso ML, Yool AJ. Mechanisms of aquaporin-facilitated cancer invasion and metastasis. Front Chem. 2018; 6: 135.

8. Wang L, Zhang Y, Wu X, Yu G. Aquaporins: new targets for cancer therapy. Technol cancer res treat. 2016; 15: 821-8.

9. Moosavi M-S, Elham Y. Aquaporins 1, 3 and 5 in different tumors, their expression, prognosis value and role as new therapeutic targets. Pathol Oncol Res. 2020; 26: 615-25.

10. Ala M, Mohammad Jafari R, Hajiabbasi A, Dehpour AR. Aquaporins and diseases pathogenesis: From trivial to undeniable involvements, a disease-based point of view. J Cell Physiol. 2021; 236: 6115-35.

11. Verkman AS, Anderson MO, Papadopoulos MC. Aquaporins: important but elusive drug targets. Nat Rev Drug discov. 2014; 13: 259-77.

12. Papadopoulos MC, Saadoun S. Key roles of aquaporins in tumor biology. Biochim et Biophys Acta. 2015; 1848: 2576-83.

13. Direito I, Madeira A, Brito MA, Soveral G. Aquaporin-5: from structure to function and dysfunction in cancer. Cell Mol Life Sci. 2016; 73: 1623-40.

14. Adeoye A, Odugbemi A, Ajewole T. Structure and function of aquaporins: the membrane water channel proteins. Biointerface Res Appl Chem. 2021; 12: 690-705.

15. Murata K, Mitsuoka K, Hirai T, Walz T, Agre P, Heymann JB, et al. Structural determinants of water permeation through aquaporin-1. Nature. 2000; 407: 599-605.

16. Dingwell DA, Brown LS, Ladizhansky V. Structure of the functionally important extracellular loop C of human aquaporin 1 obtained by solid-state NMR under nearly physiological conditions. J Phy Chem B. 2019; 123: 7700-10.

17. Frick A, Eriksson UK, de Mattia F, Öberg F, Hedfalk K, Neutze R, et al. X-ray structure of human aquaporin 2 and its implications for nephrogenic diabetes insipidus and trafficking. Proc Natl Acad Sci U S A. 2014; 111: 6305-10.

18. Ho JD, Yeh R, Sandstrom A, Chorny I, Harries WE, Robbins RA, et al. Crystal structure of human aquaporin 4 at 1.8 Å and its mechanism of conductance. Proc Natl Acad Sci U S A. 2009; 106: 7437-42.

19. Horsefield R, Nordén K, Fellert M, Backmark A, Törnroth-Horsefield S, van Scheltinga ACT, et al. High-resolution x-ray structure of human aquaporin 5. Proc Natl Acad Sci U S A. 2008; 105: 13327-32.

20. de Mare SW, Venskutonyt? R, Eltschkner S, de Groot BL, Lindkvist Petersson K. Structural basis for glycerol efflux and selectivity of human aquaporin 7. Structure. 2020; 28: 215-22.

21. Gotfryd K, Mósca AF, Missel JW, Truelsen SF, Wang K, Spulber M, et al. Human adipose glycerol flux is regulated by a pH gate in AQP10. Nature communications. 2018; 9: 1-11.

22. Verkman A, Mitra AK. Structure and function of aquaporin water channels. Am J Physiol Renal Physiol. 2000; 278: F13-F28.

23. de Groot BL, Engel A, Grubmüller H. The structure of the aquaporin-1 water channel: a comparison between cryo-electron microscopy and X-ray crystallography. J mol biol. 2003; 325: 485-93.

24. Kozono D, Yasui M, King LS, Agre P. Aquaporin water channels: atomic structure molecular dynamics meet clinical medicine. J Clin Invest. 2002; 109: 1395-9.

25. Wang F, FENG Xc, LI Ym, Yang H, MA Th. Aquaporins as potential drug targets. Acta Pharmacol Sin. 2006; 27: 395-401.

26. Benga G. On the definition, nomenclature and classification of water channel proteins (aquaporins and relatives). Mol Aspects Med. 2012; 33: 514-7.

27. Gorin MB, Yancey SB, Cline J, Revel JP, Horwitz J. The major intrinsic protein (MIP) of the bovine lens fiber membrane: characterization and structure based on cDNA cloning. Cell. 1984; 39: 49-59. 

28. Berry V, Francis P, Kaushal S, Moore A, Bhattacharya S. Missense mutations in MIP underlie autosomal dominant ‘polymorphic’and lamellar cataracts linked to 12q. Nat Genet. 2000; 25(1): 215-7.

29. Morelle J, Marechal C, Yu Z, Debaix H, Corre T, Lambie M, et al. AQP1 promoter variant, water transport, and outcomes in peritoneal dialysis. N Engl J Med. 2021; 385: 1570-80.

30. Bichet DG. Aquaporin-1 Expression and Ultrafiltration of the Peritoneal Membrane. N EnglJ Med.2021; 385: 1617-1619.

31. Chen Y, Tachibana O, Oda M, Xu R, Hamada J-i, Yamashita J, et al. Increased expression of aquaporin 1 in human hemangioblastomas and its correlation with cyst formation. J Neurooncol. 2006; 80: 219 25.

32. Endo M, Jain RK, Witwer B, Brown D. Water channel (aquaporin 1) expression and distribution in mammary carcinomas and glioblastomas. Microvas Res. 1999; 58: 89-98.

33. Hoque MO, Soria J-C, Woo J, Lee T, Lee J, Jang SJ, et al. Aquaporin 1 is overexpressed in lung cancer and stimulates NIH-3T3 cell proliferation and anchorage-independent growth. Am J Pathol. 2006; 168: 1345-53.

34. Longatti P, Basaldella L, Orvieto E, Dei Tos A, Martinuzzi A. Aquaporin (s) expression in choroid plexus tumours. Pediatr Neurosurg. 2006; 42: 228-33.

35. Tarawneh N, Hamadneh L, Abu-Irmaileh B, Shraideh Z, Bustanji Y, Abdalla S. Berberine Inhibited Growth and Migration of Human Colon Cancer Cell Lines by Increasing Phosphatase and Tensin and Inhibiting Aquaporins 1, 3 and 5 Expressions. Molecules. 2023; 28: 3823.

36. Traberg-Nyborg L, Login FH, Edamana S, Tramm T, Borgquist S, Nejsum LN. Aquaporin-1 in breast cancer. APMIS. 2022; 130: 3-10.

37. Saadoun S, Papadopoulos M, Davies D, Bell B, Krishna S. Increased aquaporin 1 water channel expression inhuman brain tumours. Br J of Cancer. 2002; 87: 621-3.

38. Vacca A, Frigeri A, Ribatti D, Nicchia GP, Nico B, Ria R, et al. Microvessel overexpression of aquaporin 1 parallels bone marrow angiogenesis in patients with active multiple myeloma. Br J Haematol. 2001; 113: 415-21.

39. El Hindy N, Bankfalvi A, Herring A, Adamzik M, Lambertz N, Zhu Y, et al. Correlation of aquaporin-1 water channel protein expression with tumor angiogenesis in human astrocytoma. Anticancer Res. 2013; 33: 609-13.

40. Niu D, Bai Y, Yao Q, Zhou L, Huang X, Zhao C. AQP2 as a diagnostic immunohistochemical marker for pheochromocytoma and/or paraganglioma. Gland Surg. 2020; 9: 200-208.

41. Wan S, Jiang J, Zheng C, Wang N, Zhai X, Fei X, et al. Estrogen nuclear receptors affect cell migration by altering sublocalization of AQP2 in glioma cell lines. Cell Death Discov. 2018; 4: 49.

42. Verkman A. Aquaporins in clinical medicine. Annu Rev Med. 2012; 63: 303-16.

43. Marlar S, Jensen HH, Login FH, Nejsum LN. Aquaporin-3 in cancer. Int J Mol Sci. 2017; 18: 2106.

44. Vandebroek A, Yasui M. Regulation of AQP4 in the central nervous system. Int JMol Sci. 2020; 21: 1603.

45. Saadoun S, Papadopoulos M, Davies D, Krishna S, Bell B. Aquaporin-4 expression is increased in oedematous human brain tumours. J Neurol Neurosurg Psychiatry. 2002; 72: 262-5.

46. Zhang Z, Chen Z, Song Y, Zhang P, Hu J, Bai C. Expression of aquaporin 5 increases proliferation and metastasis potential of lung cancer. J Pathol. 2010; 221: 210-20.

47. Bystrup M, Login FH, Edamana S, Borgquist S, Tramm T, Kwon TH, et al. Aquaporin-5 in breast cancer. APMIS. 2022; 130: 253-60.

48. Yan C, Yang J, Shen L, Chen X. Inhibitory effect of Epigallocatechin gallate on ovarian cancer cell proliferation associated with aquaporin 5 expression. Arch Gynecol Obstet. 2012; 285: 459-67.

49. Kang BW, Kim JG, Lee SJ, Chae YS, Jeong JY, Yoon GS, et al. Expression of aquaporin-1, aquaporin-3, and aquaporin-5 correlates with nodal metastasis in colon cancer. Oncology. 2015; 88: 369-76.

50. Ma J, Zhou C, Yang J, Ding X, Zhu Y, Chen X. Expression of AQP6 and AQP8 in epithelial ovarian tumor. J Mol Histol. 2016; 47: 129-34.

51. Ikeda M, Beitz E, Kozono D, Guggino WB, Agre P, Yasui M. Characterization of aquaporin-6 as a nitrate channel in mammalian cells: requirement of pore-lining residue threonine 63. J Biol Chem. 2002; 277: 39873-9.

52. Lebeck J. Metabolic impact of the glycerol channels AQP7 and AQP9 in adipose tissue and liver. J Mol Endocrinol. 2014; 52: R165-R78.

53. Dai C, Charlestin V, Wang M, Walker ZT, Miranda-Vergara MC, Facchine BA, et al. Aquaporin-7 regulates the response to cellular stress in breast cancer. Cancer Res. 2020; 80: 4071-86.

54. Shi Y-H, Chen R, Talafu T, Nijiati R, Lalai S. Significance and expression of aquaporin 1, 3, 8 in cervical carcinoma in Xinjiang Uygur women of China. Asian Pac J Cancer Prev. 2012; 13: 1971-5.

55. Zhu L, Ma N, Wang B, Wang L, Zhou C, Yan Y, et al. Significant prognostic values of aquaporin mRNA expression in breast cancer. Cancer manag Res. 2019; 1503-15.

56. Wang W, Li Q, Yang T, Bai G, Li D, Li Q, et al. Expression of AQP5 and AQP8 in human colorectal carcinoma and their clinical significance. World J Surg Oncol. 2012; 10: 1-5.

57. Yang J, Yan C, Chen X, Zhu Y. Expression of aquaglyceroporins in epithelial ovarian tumours and their clinical significance. J Int Med Res. 2011; 39: 702-11.

58. Gena P, Pellegrini-Calace M, Biasco A, Svelto M, Calamita G. Aquaporin membrane channels: biophysics, classification, functions, and possible biotechnological applications. Food Biophys. 2011; 6: 241-9.

59. Shi Z, Zhang T, Luo L, Zhao H, Cheng J, Xiang J, et al. Aquaporins in human breast cancer: identification and involvement in carcinogenesis of breast cancer. J Surg Oncol. 2012; 106: 267-72.

60. Zhang M, Li T, Zhu J, Tuo B, Liu XJJoC. Physiological and pathophysiological role of ion channels and transporters in the colorectum and colorectal cancer.J Cell Mol Med. 2020; 24: 9486-94.

61. Nico B, Ribatti DJEcr. Role of aquaporins in cell migration and edema formation in human brain tumors.Exp Cell Res. 2011; 317: 2391-6.

62. Jiang YJIl. Aquaporin-1 activity of plasma membrane affects HT20 colon cancer cell migration. IUBMB Life. 2009; 61: 1001-9.

63. Dorward HS, Du A, Bruhn MA, Wrin J, Pei JV, Evdokiou A, et al. Pharmacological blockade of aquaporin-1 water channel by AqB013 restricts migration and invasiveness of colon cancer cells and prevents endothelial tube formation in vitro. J Exp Clin Cancer Res. 2016; 35: 1-9.

64. Yoshida T, Hojo S, Sekine S, Sawada S, Okumura T, Nagata T, et al. Expression of aquaporin-1 is a poor prognostic factor for stage II and III colon cancer. Mol clin Oncol. 2013;1:953-8.

65. Moon C, Soria J-C, Jang SJ, Lee J, Hoque MO, Sibony M, et al. Involvement of aquaporins in colorectal carcinogenesis. Oncogene. 2003; 22: 6699-703.

66. Smith E, Tomita Y, Palethorpe HM, Howell S, Nakhjavani M Townsend AR, et al. Reduced aquaporin-1 transcript expression in colorectal carcinoma is associated with promoter hypermethylation. Epigenetics. 2019; 14: 158-70.

67. Li A, Lu D, Zhang Y, Li J, Fang Y, Li F, et al. Critical role of aquaporin-3 in epidermal growth factor-induced migration of colorectal carcinoma cells and its clinical significance. Oncol Rep. 2013; 29: 535-40.

68. Woo J, Lee J, Chae YK, Kim MS, Baek JH, Park JC, et al. Overexpression of AQP5, a putative oncogene, promotes cell growth and transformation. Cacer Lett. 2008; 264: 54-62.

69. Kang SK, Chae YK, Woo J, Kim MS, Park JC, Lee J, et al. Role of human aquaporin 5 in colorectal carcinogenesis. Am J Pathol. 2008; 173: 518-25.

70. Wang J, Feng L, Zhu Z, Zheng M, Wang D, Chen Z, et al. Aquaporins as diagnostic and therapeutic targets in cancer: how far we are?. J Transl Med. 2015; 13: 1-11.

71. Shan T, Cui X, Li W, Lin W, Li Y. AQP5: a novel biomarker that predicts poor clinical outcome in colorectal cancer. Oncol Rep. 2014; 32: 1564-70.

72. Wang W, Li Q, Yang T, Li D, Ding F, Sun H, et al. Anti-cancer effect of Aquaporin 5 silencing in colorectal cancer cells in association with inhibition of Wnt/β-catenin pathway. Cytotechnology. 2018; 70: 615 24.

73. Wang W, Li Q, Yang T, Li D, Ding F, Sun H, et al. RNA interference mediated silencing of aquaporin (AQP)-5 hinders angiogenesis of colorectal tumor by suppressing the production of vascular endothelial growth factor.Neoplasma. 2018; 65: 55-65.

74. Shi X, Wu S, Yang Y, Tang L, Wang Y, Dong J, et al. AQP5 silencing suppresses p38 MAPK signaling and improves drug resistance in colon cancer cells. Tumor Biol. 2014; 35: 7035-45.

75. Jensen HH, Login FH, Koffman JS, Kwon T-H, Nejsum LNJTijob. The role of aquaporin-5 in cancer cell migration: A potential active participant.Int J Biochem Cell Biol. 2016; 79: 271-6.

76. De Qing Wu ZFY, Wang KJ, Feng XY, Lv ZJ, Li Y, Jian ZXJAjocr. AQP8 inhibits colorectal cancer growth and metastasis by down-regulating PI3K/AKT signaling and PCDH7 expression. Am J Cancer Res. 2018; 8: 266.

77. Liu Y, Gao Q, Feng X, Chen G, Jiang X, Chen D, et al. Aquaporin 9 is involved in CRC metastasis through DVL2-dependent Wnt/β-catenin signaling activation. Gastroenterol Rep. 2023; 11: goad033.

78. Dajani S, Saripalli A, Sharma-Walia N. Water transport proteins aquaporins (AQPs) in cancer biology. Oncotarget. 2018; 9: 36392 36405.

79. Lan Y-L, Wang X, Lou J-C, Ma X-C, Zhang B. The potential roles of aquaporin 4 in malignant gliomas. Oncotarget. 2017; 8: 32345 32355.

80. Song T, Yang H, Ho JCM, Tang SCW, Sze SCW, Lao L, et al. Expression of aquaporin 5 in primary carcinoma and lymph node metastatic carcinoma of non-small cell lung cancer. Oncol Lett. 2015; 9: 2799 804.

81. Kitchen P, Öberg F, Sjöhamn J, Hedfalk K, Bill RM, Conner AC, et al. Plasma membrane abundance of human aquaporin 5 is dynamically regulated by multiple pathways. PloS one. 2015; 10: e0143027.

82. Beitz E, Golldack A, Rothert M, von Buelow J. Challenges and achievements in the therapeutic modulation of aquaporin functionality. Pharmacol Ther. 2015; 155: 22-35.

83. Sugimoto T, Huang L, Minematsu T, Yamamoto Y, Asada M, Nakagami G, et al. Impaired aquaporin 3 expression in reepithelialization of cutaneous wound healing in the diabetic rat. Biol Res Nurs. 2013; 15: 347-55.

84. Rodríguez A, Catalán V, Gómez-Ambrosi J, Frühbeck G. Aquaglyceroporins serve as metabolic gateways in adiposity and insulin resistance control. Cell Cycle. 2011;10:1548-56.

85. Ribatti D, Ranieri G, Annese T, Nico B. Aquaporins in cancer. Biochim Biophys Acta. 2014; 1840: 1550-3.

86. Aikman B, De Almeida A, Meier-Menches SM, Casini A. Aquaporins in cancer development: Opportunities for bioinorganic chemistry to contribute novel chemical probes and therapeutic agents. Metallomics. 2018; 10: 696-712.

87. Laforenza U, Bottino C, Gastaldi G. Mammalian aquaglyceroporin function in metabolism. Biochim Biophys Acta. 2016; 1858: 1-11.

88. Hara M, Verkman A. Glycerol replacement corrects defective skin hydration, elasticity, and barrier function in aquaporin-3-deficient mice. Proc Natl Acad Sci U S A. 2003; 100: 7360-5.

89. Matsuzaki T, Suzuki T, Koyama H, Tanaka S, Takata K. Water channel protein AQP3 is present in epithelia exposed to the environment of possible water loss. Journal of Histochem Cytochem. 1999; 47: 1275 86.

90. Li Z, Li B, Zhang L, Chen L, Sun G, Zhang Q, et al. The proliferation impairment induced by AQP3 deficiency is the result of glycerol uptake and metabolism inhibition in gastric cancer cells. Tumor Biol. 2016; 37: 9169-79.

91. Cairns R, Harris I, McCracken S, Mak T, editors. Cancer cell metabolism. Cold Spring Harb Symp Quant Biol. 2011; 76: 299-311.

92. Boroughs LK, DeBerardinis RJ. Metabolic pathways promoting cancer cell survival and growth. Nat Cell Biol. 2015;17:351-9.

93. Watt MJ, Steinberg GR. Regulation and function of triacylglycerol lipases in cellular metabolism. Biochem J. 2008; 414: 313-25.

94. Verkman A, Hara-Chikuma M, Papadopoulos MC. Aquaporins—new players in cancer biology. Journal of Mol Med. 2008; 86: 523-9.

95. Hara-Chikuma M, Verkman A. Prevention of skin tumorigenesis and impairment of epidermal cell proliferation by targeted aquaporin-3 gene disruption. Mol Cell Biol. 2008; 28: 326-32.

96. Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011; 11: 325-37.

97. Pavlova NN, Thompson CB. The emerging hallmarks of cancer metabolism. Cell Metab. 2016; 23: 27-47.

98. Pike Winer LS, Wu M. Rapid analysis of glycolytic and oxidative substrate flux of cancer cells in a microplate. PloS One. 2014; 9: e109916.

99. Warth A, Mittelbronn M, Hülper P, Erdlenbruch B, Wolburg H. Expression of the water channel protein aquaporin-9 in malignant brain tumors. Appl Immunohistochem Mole Morphol. 2007;15:193 8.

100. Tan G, Sun S, Yuan D. Expression of the water channel protein aquaporin-9 in human astrocytic tumours: correlation with pathological grade. J Int Med Res. 2008; 36: 777-82.

101. Zhang W-g, Li C-f, Liu M, Chen X-f, Shuai K, Kong X, et al. Aquaporin 9 is down-regulated in hepatocellular carcinoma and its over expression suppresses hepatoma cell invasion through inhibiting epithelial-to-mesenchymal transition. Cancer Lett. 2016; 378: 111 9.

102. Schinkel AH, Jonker JW. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview. Adv Drug Deliv Rev. 2012; 64: 138-53.

103. van Zanden JJ, Geraets L, Wortelboer HM, van Bladeren PJ, Rietjens IM, Cnubben NH. Structural requirements for the flavonoid mediated modulation of glutathione S-transferase P1-1 and GS-X pump activity in MCF7 breast cancer cells. Biochem Pharmacol. 2004; 67: 1607-17.

104. Oloumi A, MacPhail SH, Johnston PJ, Banáth JP, Olive PL. Changes in subcellular distribution of topoisomerase IIα correlate with etoposide resistance in multicell spheroids and xenograft tumors. Cancer Res. 2000; 60: 5747-53.

105. Beck WT. The cell biology of multiple drug resistance. Biochem Pharmacol. 1987;36:2879-87.

106. Gao L, Gao Y, Li X, Howell P, Kumar R, Su X, et al. Aquaporins mediate the chemoresistance of human melanoma cells to arsenite. Mol Oncol. 2012; 6: 81-7.

107. Chae YK, Kang SK, Kim MS, Woo J, Lee J, Chang S, et al. Human AQP5 plays a role in the progression of chronic myelogenous leukemia (CML). PLoS One. 2008; 3: e2594.

108. Guo X, Ma N, Wang J, Song J, Bu X, Cheng Y, et al. Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells. BMC Cancer. 2008; 8: 1-9.

109. Shen H, Xu W, Luo W, Zhou L, Yong W, Chen F, et al. Upregulation of mdr1 gene is related to activation of the MAPK/ERK signal transduction pathway and YB-1 nuclear translocation in B-cell lymphoma. Exp Hematol. 2011; 39: 558-69.

110. Sui H, Fan Z, Li Q. Signal transduction pathways and transcriptional mechanisms of ABCB1/Pgp-mediated multiple drug resistance in human cancer cells. J Int Med Res. 2012; 40: 426-35.

111. Tomiyasu H, Watanabe M, Goto-Koshino Y, Fujino Y, Ohno K, Sugano S, et al. Regulation of expression of ABCB1 and LRP genes by mitogen-activated protein kinase/extracellular signal-regulated kinase pathway and its role in generation of side population cells in canine lymphoma cell lines. Leuk Lymphoma. 2013; 54: 1309-15.

112. Galoian K, Temple H, Galoyan A. mTORC1 inhibition and ECM–cell adhesion-independent drug resistance via PI3K–AKT and PI3K RAS–MAPK feedback loops. Tumor Biol. 2012; 33: 885-90.

113. Zhang W, Liu HT. MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res. 2002; 12: 9-18.

114. Bienert GP, Møller AL, Kristiansen KA, Schulz A, Møller IM, Schjoerring JK, et al. Specific aquaporins facilitate the diffusion of hydrogen peroxide across membranes. J Biol Chem. 2007; 282: 1183-92.

115. Rodrigues C, Mósca AF, Martins AP, Nobre T, Prista C, Antunes F, et al. Rat aquaporin-5 is pH-gated induced by phosphorylation and is implicated in oxidative stress. Int J Mol Sci. 2016; 17: 2090.

116. Rodrigues C, Pimpão C, Mósca AF, Coxixo AS, Lopes D, da Silva IV, et al. Human aquaporin-5 facilitates hydrogen peroxide permeation affecting adaption to oxidative stress and cancer cell migration. Cancers. 2019; 11: 932.

117. Prata C, Hrelia S, Fiorentini D. Peroxiporins in cancer. Int J Mol Sci. 2019; 20: 1371.

118. ?ipak Gašparovi? A, Milkovi? L, Rodrigues C, Mlinari? M, Soveral G. Peroxiporins Are Induced upon Oxidative Stress Insult and Are Associated with Oxidative Stress Resistance in Colon Cancer Cell Lines. Antioxidants. 2021; 10: 1856.

119. Jablonski EM, Mattocks MA, Sokolov E, Koniaris LG, Hughes Jr FM, Fausto N, et al. Decreased aquaporin expression leads to increased resistance to apoptosis in hepatocellular carcinoma. Cancer Lett. 2007; 250: 36-46.

120. Yang B, Zhao D, Verkman A. Evidence against functionally significant aquaporin expression in mitochondria. J Biol Chem. 2006; 281: 16202-6.

121. Dong X, Wang Y, Zhou Y, Wen J, Wang S, Shen L. Aquaporin 3 facilitates chemoresistance in gastric cancer cells to cisplatin via autophagy. Cell Death Discov. 2016; 2: 1-6.

122. Maes H, Rubio N, Garg AD, Agostinis P. Autophagy: shaping the tumor microenvironment and therapeutic response. Trends Mol Med. 2013; 19: 428-46.

123. Ozpolat B, Benbrook DM. Targeting autophagy in cancer management–strategies and developments. Cancer Manag Res. 2015; 7: 291-9.

124. Ojha R, Bhattacharyya S, Singh SK. Autophagy in cancer stem cells: a potential link between chemoresistance, recurrence, and metastasis. BioRes Open Access. 2015; 4: 97-108.

125. Li X, Pei B, Wang H, Tang C, Zhu W, Jin F. Effect of AQP-5 silencing by siRNA interference on chemosensitivity of breast cancer cells. OncoTargets Ther. 2018; 11: 3359-3369.

126. Imaizumi H, Ishibashi K, Takenoshita S, Ishida H. Aquaporin 1 expression is associated with response to adjuvant chemotherapy in stage II and III colorectal cancer. Oncology Lett. 2018; 15: 6450-6.

127. Chong W, Zhang H, Guo Z, Yang L, Shao Y, Liu X, et al. Aquaporin 1 promotes sensitivity of anthracycline chemotherapy in breast cancer by inhibiting β-catenin degradation to enhance TopoIIα activity. Cell Death Differ. 2021; 28: 382-400.

128. Zhu Z, Jiao L, Li T, Wang H, Wei W, Qian H. Expression of AQP3 and AQP5 as a prognostic marker in triple-negative breast cancer. Oncology Lett. 2018; 16: 2661-7.

129. Li J, Wang Z, Chong T, Chen H, Li H, Li G, et al. Over-expression of a poor prognostic marker in prostate cancer: AQP5 promotes cells growth and local invasion. World J Surg Oncol. 2014; 12: 1-9. 

130. Morrissey JJ, Mellnick VM, Luo J, Siegel MJ, Figenshau RS, Bhayani S, et al. Evaluation of Urine Aquaporin-1 and Perilipin-2 Concentrations as Biomarkers to Screen for Renal Cell Carcinoma: A Prospective Cohort Study. JAMA Oncol. 2015; 1: 204-12.

131. Imrédi E, Tóth B, Doma V, Barbai T, Rásó E, Kenessey I, et al. Aquaporin 1 protein expression is associated with BRAF V600 mutation and adverse prognosis in cutaneous melanoma. Melanoma Res. 2016; 26: 254-60.

132. Shan T, Chen S, Chen X, Kong D, Lin W, Li W, et al. Impact of AQP-5 on the growth of colorectal cancer cells and the underlying mechanism. Int J Clin Exp Pathol. 2018; 11: 58-67.

133. Kang BW, Kim JG, Chae YS, Lee SJ, Sohn SK, Moon JH, et al. AQP1 expression and survival in patients with colon cancer. J Clin Oncol. 2014; 32: e14586-e.

134. Gao C, Shen J, Yao L, Xia Z, Liang X, Zhu R, et al. Low expression of AQP9 and its value in hepatocellular carcinoma. Transl Cancer Res. 2021; 10: 1826-41.

135. Shan T, Zheng B, Chen X, Tao W, Erli J, Bai Y, et al. Expression of AQP5 in colorectal cancer and its relationship with clinical outcome. J Xi’an Jiaotong Univ (Med.Sci). 2015: 815-8.

136. Abdelrahman AE, Ibrahim DA, El-Azony A, Alnagar AA, Ibrahim A. ERCC1, PARP-1, and AQP1 as predictive biomarkers in colon cancer patients receiving adjuvant chemotherapy. Cancer Biomark. 2020; 27: 251-64.

137. Dou R, Deng Y, Huang L, Fu S, Tan S, Wang L, et al. Multi-microarray identifies lower AQP9 expression in adjuvant chemotherapy nonresponders with stage III colorectal cancer. Cancer Lett. 2013;336:106-13.

138. Wang Y, Fan Y, Zheng C, Zhang X. Knockdown of AQP1 inhibits growth and invasion of human ovarian cancer cells. Mol Med Rep. 2017; 16: 5499-504.

139. Zhang Q, Lin L, Li W, Lu G, Li X. MiR-223 inhibitor suppresses proliferation and induces apoptosis of thyroid cancer cells by down regulating aquaporin-1. J Recept Signal Transduct Res. 2019;39:146 53.

140. Nakhjavani M, Palethorpe HM, Tomita Y, Smith E, Price TJ, Yool AJ, et al. Stereoselective anti-cancer activities of ginsenoside Rg3 on triple negative breast cancer cell models. Pharmaceuticals. 2019; 12: 117.

141. Pan X-Y, Guo H, Han J, Hao F, An Y, Xu Y, et al. Ginsenoside Rg3 attenuates cell migration via inhibition of aquaporin 1 expression in PC-3M prostate cancer cells. Eur J pharmacol. 2012; 683: 27-34.

142. Kong B, Zhao S, Kang X, Wang B. MicroRNA-133a-3p inhibits cell proliferation, migration and invasion in colorectal cancer by targeting AQP1. Oncology Lett. 2021; 22: 1-10.

143. Kourghi M, Pei JV, De Ieso ML, Flynn G, Yool AJ. Bumetanide derivatives AqB007 and AqB011 selectively block the aquaporin-1 ion channel conductance and slow cancer cell migration. Mol pharmacol. 2016; 89: 133-40.

144. Zhang X, Chen Y, Dong L, Shi B. Effect of selective inhibition of aquaporin 1 on chemotherapy sensitivity of J82 human bladder cancer cells. Oncology Lett. 2018; 15: 3864-9.

145. Bin K, Shi-Peng Z. Acetazolamide inhibits aquaporin-1 expression and colon cancer xenograft tumor growth. Hepatogastroenterology. 2011;58:1502-6.

146. De Ieso ML, Pei JV, Nourmohammadi S, Smith E, Chow PH, Kourghi M, et al. Combined pharmacological administration of AQP1 ion channel blocker AqB011 and water channel blocker Bacopaside II amplifies inhibition of colon cancer cell migration. Sci Rep. 2019; 9: 12635.

147. Fathizadeh H, Asemi Z. Epigenetic roles of PIWI proteins and piRNAs in lung cancer. Cell Biosci. 2019; 9: 1-8.

148. Fathizadeh H, Hallajzadeh J, Asemi Z. Circular RNAs as diagnostic biomarker in pancreatic cancer. Pathol Res Pract. 2020; 216: 153075.

149. Jafari D, Noorbakhsh F, Delavari A, Tavakkoli-Bazzaz J, Farashi Bonab S, Abdollahzadeh R, et al. Expression level of long noncoding RNA NKILAmiR103-miR107 inflammatory axis and its clinical significance as potential biomarker in patients with colorectal cancer. J Res Med Sci. 2020; 25.

150. Chu Y, Wang X, Yu N, Li Y, Kan J. Long non-coding RNA FGD5-AS1/ microRNA-133a-3p upregulates aquaporin 1 to decrease the inflammatory response in LPS-induced sepsis. Mol Med Rep. 2021; 24: 1-11.

151. Nave M, Castro RE, Rodrigues CM, Casini A, Soveral G, Gaspar MM. Nanoformulations of a potent copper-based aquaporin inhibitor with cytotoxic effect against cancer cells. Nanomedicine. 2016; 11: 1817-30.

152. Chen C, Ma T, Zhang C, Zhang H, Bai L, Kong L, et al. Down-regulation of aquaporin 5-mediated epithelial-mesenchymal transition and anti-metastatic effect by natural product Cairicoside E in colorectal cancer. Mol Carcinog. 2017; 56: 2692-705.

153. Li Q, Yang T, Li D, Ding F, Bai G, Wang W, et al. Knockdown of aquaporin-5 sensitizes colorectal cancer cells to 5-fluorouracil via inhibition of the Wnt–β-catenin signaling pathway. Biochem Cell Biol. 2018; 96: 572-9.

Mohammadabadi MA, Moazzeni A, Jafarzadeh L, Faraji F, et al. (2023) Aquaporins in Colorectal Cancer: Exploring Their Role in Tumorigenesis, Metastasis, and Drug Response. J Cancer Biol Res 10(2): 1140.

Received : 15 Sep 2023
Accepted : 31 Oct 2023
Published : 31 Oct 2023
Journals
Annals of Otolaryngology and Rhinology
ISSN : 2379-948X
Launched : 2014
JSM Schizophrenia
Launched : 2016
Journal of Nausea
Launched : 2020
JSM Internal Medicine
Launched : 2016
JSM Hepatitis
Launched : 2016
JSM Oro Facial Surgeries
ISSN : 2578-3211
Launched : 2016
Journal of Human Nutrition and Food Science
ISSN : 2333-6706
Launched : 2013
JSM Regenerative Medicine and Bioengineering
ISSN : 2379-0490
Launched : 2013
JSM Spine
ISSN : 2578-3181
Launched : 2016
Archives of Palliative Care
ISSN : 2573-1165
Launched : 2016
JSM Nutritional Disorders
ISSN : 2578-3203
Launched : 2017
Annals of Neurodegenerative Disorders
ISSN : 2476-2032
Launched : 2016
Journal of Fever
ISSN : 2641-7782
Launched : 2017
JSM Bone Marrow Research
ISSN : 2578-3351
Launched : 2016
JSM Mathematics and Statistics
ISSN : 2578-3173
Launched : 2014
Journal of Autoimmunity and Research
ISSN : 2573-1173
Launched : 2014
JSM Arthritis
ISSN : 2475-9155
Launched : 2016
JSM Head and Neck Cancer-Cases and Reviews
ISSN : 2573-1610
Launched : 2016
JSM General Surgery Cases and Images
ISSN : 2573-1564
Launched : 2016
JSM Anatomy and Physiology
ISSN : 2573-1262
Launched : 2016
JSM Dental Surgery
ISSN : 2573-1548
Launched : 2016
Annals of Emergency Surgery
ISSN : 2573-1017
Launched : 2016
Annals of Mens Health and Wellness
ISSN : 2641-7707
Launched : 2017
Journal of Preventive Medicine and Health Care
ISSN : 2576-0084
Launched : 2018
Journal of Chronic Diseases and Management
ISSN : 2573-1300
Launched : 2016
Annals of Vaccines and Immunization
ISSN : 2378-9379
Launched : 2014
JSM Heart Surgery Cases and Images
ISSN : 2578-3157
Launched : 2016
Annals of Reproductive Medicine and Treatment
ISSN : 2573-1092
Launched : 2016
JSM Brain Science
ISSN : 2573-1289
Launched : 2016
JSM Biomarkers
ISSN : 2578-3815
Launched : 2014
JSM Biology
ISSN : 2475-9392
Launched : 2016
Archives of Stem Cell and Research
ISSN : 2578-3580
Launched : 2014
Annals of Clinical and Medical Microbiology
ISSN : 2578-3629
Launched : 2014
JSM Pediatric Surgery
ISSN : 2578-3149
Launched : 2017
Journal of Memory Disorder and Rehabilitation
ISSN : 2578-319X
Launched : 2016
JSM Tropical Medicine and Research
ISSN : 2578-3165
Launched : 2016
JSM Head and Face Medicine
ISSN : 2578-3793
Launched : 2016
JSM Cardiothoracic Surgery
ISSN : 2573-1297
Launched : 2016
JSM Bone and Joint Diseases
ISSN : 2578-3351
Launched : 2017
JSM Bioavailability and Bioequivalence
ISSN : 2641-7812
Launched : 2017
JSM Atherosclerosis
ISSN : 2573-1270
Launched : 2016
Journal of Genitourinary Disorders
ISSN : 2641-7790
Launched : 2017
Journal of Fractures and Sprains
ISSN : 2578-3831
Launched : 2016
Journal of Autism and Epilepsy
ISSN : 2641-7774
Launched : 2016
Annals of Marine Biology and Research
ISSN : 2573-105X
Launched : 2014
JSM Health Education & Primary Health Care
ISSN : 2578-3777
Launched : 2016
JSM Communication Disorders
ISSN : 2578-3807
Launched : 2016
Annals of Musculoskeletal Disorders
ISSN : 2578-3599
Launched : 2016
Annals of Virology and Research
ISSN : 2573-1122
Launched : 2014
JSM Renal Medicine
ISSN : 2573-1637
Launched : 2016
Journal of Muscle Health
ISSN : 2578-3823
Launched : 2016
JSM Genetics and Genomics
ISSN : 2334-1823
Launched : 2013
JSM Anxiety and Depression
ISSN : 2475-9139
Launched : 2016
Clinical Journal of Heart Diseases
ISSN : 2641-7766
Launched : 2016
Annals of Medicinal Chemistry and Research
ISSN : 2378-9336
Launched : 2014
JSM Pain and Management
ISSN : 2578-3378
Launched : 2016
JSM Women's Health
ISSN : 2578-3696
Launched : 2016
Clinical Research in HIV or AIDS
ISSN : 2374-0094
Launched : 2013
Journal of Endocrinology, Diabetes and Obesity
ISSN : 2333-6692
Launched : 2013
Journal of Substance Abuse and Alcoholism
ISSN : 2373-9363
Launched : 2013
JSM Neurosurgery and Spine
ISSN : 2373-9479
Launched : 2013
Journal of Liver and Clinical Research
ISSN : 2379-0830
Launched : 2014
Journal of Drug Design and Research
ISSN : 2379-089X
Launched : 2014
JSM Clinical Oncology and Research
ISSN : 2373-938X
Launched : 2013
JSM Bioinformatics, Genomics and Proteomics
ISSN : 2576-1102
Launched : 2014
JSM Chemistry
ISSN : 2334-1831
Launched : 2013
Journal of Trauma and Care
ISSN : 2573-1246
Launched : 2014
JSM Surgical Oncology and Research
ISSN : 2578-3688
Launched : 2016
Annals of Food Processing and Preservation
ISSN : 2573-1033
Launched : 2016
Journal of Radiology and Radiation Therapy
ISSN : 2333-7095
Launched : 2013
JSM Physical Medicine and Rehabilitation
ISSN : 2578-3572
Launched : 2016
Annals of Clinical Pathology
ISSN : 2373-9282
Launched : 2013
Annals of Cardiovascular Diseases
ISSN : 2641-7731
Launched : 2016
Journal of Behavior
ISSN : 2576-0076
Launched : 2016
Annals of Clinical and Experimental Metabolism
ISSN : 2572-2492
Launched : 2016
Clinical Research in Infectious Diseases
ISSN : 2379-0636
Launched : 2013
JSM Microbiology
ISSN : 2333-6455
Launched : 2013
Journal of Urology and Research
ISSN : 2379-951X
Launched : 2014
Journal of Family Medicine and Community Health
ISSN : 2379-0547
Launched : 2013
Annals of Pregnancy and Care
ISSN : 2578-336X
Launched : 2017
JSM Cell and Developmental Biology
ISSN : 2379-061X
Launched : 2013
Annals of Aquaculture and Research
ISSN : 2379-0881
Launched : 2014
Clinical Research in Pulmonology
ISSN : 2333-6625
Launched : 2013
Journal of Immunology and Clinical Research
ISSN : 2333-6714
Launched : 2013
Annals of Forensic Research and Analysis
ISSN : 2378-9476
Launched : 2014
JSM Biochemistry and Molecular Biology
ISSN : 2333-7109
Launched : 2013
Annals of Breast Cancer Research
ISSN : 2641-7685
Launched : 2016
Annals of Gerontology and Geriatric Research
ISSN : 2378-9409
Launched : 2014
Journal of Sleep Medicine and Disorders
ISSN : 2379-0822
Launched : 2014
JSM Burns and Trauma
ISSN : 2475-9406
Launched : 2016
Chemical Engineering and Process Techniques
ISSN : 2333-6633
Launched : 2013
Annals of Clinical Cytology and Pathology
ISSN : 2475-9430
Launched : 2014
JSM Allergy and Asthma
ISSN : 2573-1254
Launched : 2016
Journal of Neurological Disorders and Stroke
ISSN : 2334-2307
Launched : 2013
Annals of Sports Medicine and Research
ISSN : 2379-0571
Launched : 2014
JSM Sexual Medicine
ISSN : 2578-3718
Launched : 2016
Annals of Vascular Medicine and Research
ISSN : 2378-9344
Launched : 2014
JSM Biotechnology and Biomedical Engineering
ISSN : 2333-7117
Launched : 2013
Journal of Hematology and Transfusion
ISSN : 2333-6684
Launched : 2013
JSM Environmental Science and Ecology
ISSN : 2333-7141
Launched : 2013
Journal of Cardiology and Clinical Research
ISSN : 2333-6676
Launched : 2013
JSM Nanotechnology and Nanomedicine
ISSN : 2334-1815
Launched : 2013
Journal of Ear, Nose and Throat Disorders
ISSN : 2475-9473
Launched : 2016
JSM Ophthalmology
ISSN : 2333-6447
Launched : 2013
Journal of Pharmacology and Clinical Toxicology
ISSN : 2333-7079
Launched : 2013
Annals of Psychiatry and Mental Health
ISSN : 2374-0124
Launched : 2013
Medical Journal of Obstetrics and Gynecology
ISSN : 2333-6439
Launched : 2013
Annals of Pediatrics and Child Health
ISSN : 2373-9312
Launched : 2013
JSM Clinical Pharmaceutics
ISSN : 2379-9498
Launched : 2014
JSM Foot and Ankle
ISSN : 2475-9112
Launched : 2016
JSM Alzheimer's Disease and Related Dementia
ISSN : 2378-9565
Launched : 2014
Journal of Addiction Medicine and Therapy
ISSN : 2333-665X
Launched : 2013
Journal of Veterinary Medicine and Research
ISSN : 2378-931X
Launched : 2013
Annals of Public Health and Research
ISSN : 2378-9328
Launched : 2014
Annals of Orthopedics and Rheumatology
ISSN : 2373-9290
Launched : 2013
Journal of Clinical Nephrology and Research
ISSN : 2379-0652
Launched : 2014
Annals of Community Medicine and Practice
ISSN : 2475-9465
Launched : 2014
Annals of Biometrics and Biostatistics
ISSN : 2374-0116
Launched : 2013
JSM Clinical Case Reports
ISSN : 2373-9819
Launched : 2013
Journal of Surgery and Transplantation Science
ISSN : 2379-0911
Launched : 2013
Journal of Dermatology and Clinical Research
ISSN : 2373-9371
Launched : 2013
JSM Gastroenterology and Hepatology
ISSN : 2373-9487
Launched : 2013
Annals of Nursing and Practice
ISSN : 2379-9501
Launched : 2014
JSM Dentistry
ISSN : 2333-7133
Launched : 2013
Author Information X