Next Article in Journal
Predicting Immunotherapy Outcomes in Older Patients with Solid Tumors Using the LIPI Score
Next Article in Special Issue
Combination Modality Using Quercetin to Enhance the Efficacy of Docetaxel in Prostate Cancer Cells
Previous Article in Journal
Identification of EGFR-Related LINC00460/mir-338-3p/MCM4 Regulatory Axis as Diagnostic and Prognostic Biomarker of Lung Adenocarcinoma Based on Comprehensive Bioinformatics Analysis and Experimental Validation
Previous Article in Special Issue
Prostate Cancer Tumor Stroma: Responsibility in Tumor Biology, Diagnosis and Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Obesity-Related Cross-Talk between Prostate Cancer and Peripheral Fat: Potential Role of Exosomes

1
The First Clinical College, Gannan Medical University, Ganzhou 341000, China
2
Department of Urology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
3
Institute of Urology, The First Affiliated Hospital of Ganna Medical University, Ganzhou 341000, China
4
Jiangxi Engineering Technology Research Center of Calculi Prevention, Ganzhou 341000, China
*
Authors to whom correspondence should be addressed.
These authors have contributed equally to this work and share first authorship.
Submission received: 13 September 2022 / Revised: 6 October 2022 / Accepted: 13 October 2022 / Published: 17 October 2022

Abstract

:

Simple Summary

Obesity is involved in many aspects of prostate cancer progression as a risk factor for prostate cancer, especially in the process of biochemical recurrence in the prostate. Approximately 27–53% of prostate cancer patients can develop biochemical recurrence after radical prostatectomy, which poses difficulties in the clinical management of prostate cancer, and this is closely related to the release of exosomes from adipose tissue in the obese state. In this review, we summarize the crosstalk between prostate cancer peripheral adiposity and prostate cancer and discuss the potential role of exosomes in this process and the prospects for the use of adipose exosomes. Exosomes play an important role in the crosstalk between the two this may be a new basis to explain obesity as a biochemical recurrence after prostate cancer surgery and a potential avenue for future prostate therapy.

Abstract

The molecular mechanisms of obesity-induced cancer progression have been extensively explored because of the significant increase in obesity and obesity-related diseases worldwide. Studies have shown that obesity is associated with certain features of prostate cancer. In particular, bioactive factors released from periprostatic adipose tissues mediate the bidirectional communication between periprostatic adipose tissue and prostate cancer. Moreover, recent studies have shown that extracellular vesicles have a role in the relationship between tumor peripheral adipose tissue and cancer progression. Therefore, it is necessary to investigate the feedback mechanisms between prostate cancer and periglandular adipose and the role of exosomes as mediators of signal exchange to understand obesity as a risk factor for prostate cancer. This review summarizes the two-way communication between prostate cancer and periglandular adipose and discusses the potential role of exosomes as a cross-talk and the prospect of using adipose tissue as a means to obtain exosomes in vitro. Therefore, this review may provide new directions for the treatment of obesity to suppress prostate cancer.

1. Introduction

Prostate cancer (PCa) is the most common cancer among men in developed countries. It is also one of the cancers with significantly increased incidence in developing countries [1,2]. The reasons for the increasing incidence of PCa in Asian countries year by year cannot be explained solely by an increase in the level of screening and diagnosis based on the implementation of PSA. This increased incidence of PCa also largely reflects the rise in risk factors associated with PCa. Obesity is associated with a more aggressive tumor, poorer treatment outcomes, and a higher risk of mortality for PCa patients [3,4]. Several clinical studies have indicated that a higher visceral adiposity is associated with higher grade or aggressiveness of PCa [5,6]. Various mechanistic studies of obesity-mediated cancer progression have further shown that obesity influenced the aggressiveness of PCa due to increased systemic inflammation, hyperinsulinemia, altered adipokine profile, and increased lipid function [7,8]. Although there are very few adipocytes in the prostate [9], clinical and in vitro laboratory studies have evaluated the relationship and potential mechanisms between periprostatic carcinoma adipose (PPAT) and PCa progression [10,11,12,13] (Table 1).
Obesity is associated with the incidence of different cancers, poor treatment outcomes, and high cancer-related mortality rates [29]. Although individual studies have found that the incidence of PCa, lung, colorectal, and ovarian cancer is inversely associated with patient BMI [30]. Beyond that, a significant association between obesity and PCa progression has been established, especially in relation to more malignant and biochemical recurrence (biochemical recurrence defined as patients without postoperative endocrine therapy and radiotherapy, with two consecutive prostate-specific antigen (PSA) ≥ 0.2 μg/L during follow-up) of PCa [31,32,33]. Meta-analyses have also shown that obesity is related to more advanced prostate cancer [4,34]. Cells from tissues surrounding or adjacent to prostate cancer, such as stromal cells, endothelial cells, immune cells, and adipose stem cells, can form a tumor microenvironment that promotes prostate cancer survival [35]. This also indicates that peri-Pca adipose tissue and its tumor mesenchymal cells can promote Pca growth through various mechanisms, including the release of growth factors, inflammatory signaling activators, and the release of fatty acids to provide energy for Pca. Besides the specific regulation of these biological factors, exosomes secreted by peri-Pca adipocytes may also play a key role in Pca growth. For example, changes in the amount and status of white adipose tissue (WAT) and brown adipose tissue (BAT) caused by obesity affect the amount and cargo of exosomes released into circulation. The unhealthy expansion of adipose tissue in the obese state leads to local hypoxia, which induces chronic inflammation. Adipocyte dysfunction then induces changes in exosomes [36], thus mediating PCa progression through various molecular mechanisms [37].
As a new mode of cellular communication, extracellular vesicles contain cellular molecules with various physiological roles. They can be divided into two categories based on their size: vesicles of 50–1000 nm formed via germination and exosomes of 40–160 nm from the somatic membrane [38]. Exosomes have many sources, including all living cells containing RNA, bioactive lipids, and proteins [39], with various functions related to biological processes. Exosomes participate in pathological and physiological regulatory processes by transporting these substances. Besides the pro-carcinogenic role of tumor mesenchymal adipocyte exosomes, peritumor adipose tissue may also communicate this process through exosomes. Exosomes may play a key role in the cross-talk between PCa and peri-tumor adipose tissue. However, studies have not comprehensively evaluated the PPAT phenotype and its detailed cross-talk with PCa. Furthermore, the role of exosomes in the association between PCa and peri-cancerous fat is unclear. It is also unclear whether removal of this fat and inhibition of exosome release is beneficial for patients and whether obesity exacerbates the ability of exosomes to mediate this process.

2. Cross-Talk between PCa and PPAT

PPAT is the adipose tissue surrounding the surface of the prostate (Figure 1). PPAT varies in thickness and structure at different locations in the prostate [40]. However, PCa cells communicate with adipose tissue cells in the same way (defined as extraperitoneal extension of the tumor) after PCa invasion. This process is usually associated with a significantly poorer prognosis for PCa [41,42]. Adipocytes within the omentum promote the initial homing of ovarian cancer cells to the omentum and provide fatty acids to cancer cells, thus promoting rapid tumor growth by secreting adipokines [43]. PPAT, which is external to the cancerous tissue, also plays a role in PCa progression. Studies of PPAT thickness, expression of inflammatory factors, and the involvement of obesity in prostate cancer progression have shown that changes in local adipose tissue in the prostate body can influence the behavior of PCa [13]. In addition, PCa induces pro-tumorigenesis in peripheral adipose tissue. For example, preadipocytes triggered by PCa cell-culture medium undergo tumor-like transformations, including genetic variability, epithelial–mesenchymal transition (EMT), and tumor-like lesion formation in vivo [44]. This suggests that the continuous positive feedback process between PCa and PPAT accelerates the continuous deterioration of PCa.

2.1. PPAT Promotes Survival and Progression of Prostate Cancer

Like the obesity epidemic, the understanding of adipocytes and adipose tissue is expanding. Major advances in the last decade have provided new insights into the role of adipose tissue in normal physiology and obesity-related complications. As a result, adipocyte biology studies have focused on the global metabolic disease pandemic [45]. Adipocytes can secrete various effectors, including exosomes, miRNAs, lipids, and inflammatory cytokines, which play an important role in paracrine and endocrine influencing local and systemic metabolic responses. Adipose tissue acts as an endocrine organ or embedded energy store to actively promote tumor growth and metastasis by secreting extracellular matrix components, such as adipokines, pro-inflammatory cytokines, and pro-angiogenic factors [21,46,47,48]. However, obesity exacerbates this process. A study reported that obesity is associated with a poorer PCa prognosis [49]. Obesity may lead to the development of high-grade prostate cancer by increasing aromatase activity and regulating the secretion of various cytokines such as adipokines, vascular endothelial growth factors, and prostaglandins [50,51]. However, it is unclear how the interaction and single effect of multiple factors influence the tandem process between obesity and PCa.
The main biologic factors released by adipocytes (adipokines and inflammatory factors) are recognized biological mediators of cross-talk between PPAT and PCa. Secretion of multiple biologic factors by adipocytes was associated with a significant increase in the proliferation, migration, and invasive capacity of PCa cells [21,46,52]. In one study, adipokine CCL7 overexpression was detected in prostate specimens collected from obese patients after puncture and radical PCa surgery. Adipokine CCL7 was positively correlated with PCa pathological malignancy than normal punctured prostate tissue, suggesting that PPAT can enhance the malignancy of PCa cells [52]. PPAT contains MMP (matrix metalloproteinase: plays an important role in many cellular behaviors) with higher activity than abdominal visceral adipose tissue [21]. Activated MMP can degrade extracellular matrix proteins and thus promote the invasion of cancer cells into surrounding tissues [53]. In vivo experiments also revealed that mature epididymal adipocytes from rats or humans with PCa accelerated the growth and differentiation of normal rat or human prostate epithelial cells. These are also accompanied by enhanced expression of VEGF (vascular endothelial growth factor) and PdGF (platelet-derived growth factor) in prostate epithelial cells and activation of the PI3K pathway in PC3 cells [54,55]. Adipose tissue in obese patients is always in a state of chronic inflammation, characterized by overexpression of inflammatory factors secreted by adipose tissue [56], including IL-6 (interleukin-6: cytokines involved in the inflammatory response), IL-8 (interleukin-8: cytokines involved in the inflammatory response), MCP-1 (monocyte chemoattractant protein 1), and TNF-α (tumor necrosis factor α), possibly due to excessive infiltration of immune cells, such as macrophages in obese tissues [57]. However, further studies should assess whether the degree of inflammation in PPAT is correlated with obesity or metabolic syndrome. Several in vivo and in vitro experiments have shown that these inflammatory factors are closely associated with the progression of PCa [58,59,60]. Another study also showed that the degree of inflammation in PPAT was associated with adipocyte size, insulin and triglyceride circulating levels, and high levels of PCa [61]. Furthermore, the appearance of cachexia in advanced tumor stages may be due to the metabolic changes in adipocytes caused by cancer cells, which leads to lipolytic pathway activation, making adipocytes a key energy source for cancer cells. This situation also applies to PPAT with advanced PCa. For example, the expression of lipolytic enzymes and CGI-58 was more pronounced in late PCa [62]. FTIR microspectroscopy (an optically based technique that can measure the transitions in vibrational modes of the functional groups of bio-molecular constituents within cells) also showed that lipid translocation occurs between adipocytes and PCa cells [63]. NMR spectroscopy (a technique applying nuclear magnetic resonance phenomena for the determination of molecular structure) showed that patients with high-grade PCa have higher rates of monounsaturated/saturated fatty acids than those in low-grade patients [64]. Fatty acids serve as an important energy source for cancer cells, and studying how PPAT affects the uptake and utilization of fatty acids by PCa and the detailed mechanism of action of these fatty acids on PCa may improve the understanding of the mutual cross-linking between obesity and PCa. PPAT can also interfere with the expression of hormones that promote PCa progression. PPAT, as a source of local extra-gonadal androgens, promotes the growth and metastasis of PCa. PPAT also expresses aromatase, which converts androgens to estrogens [65] and plays a crucial role in the pathogenesis and progression of PCa. For example, estrogens can activate wild-type and mutant androgen receptors, limiting ADT treatment [66]. In addition, obesity may exacerbate this process by altering the metabolic and endocrine characteristics of multiple adipose tissues, thereby increasing the release of multiple biological factors, as well as the mobilization of free fatty acids [52,67,68]. Obesity also increases the rate of preadipocyte migration in WAT, leading to obesity-induced PCa progression [69,70]. PPAT is more active in metabolism and secretion in obese men than in lean men. For example, PPAT in obese men has higher MMP-9 activity, which leads to better induction of PC3 cell lineage and endothelial cell proliferation [71]. PPAT from obese men has a higher expression of chemokine CXCL1 than that of lean men [72], which may lead to PCa susceptibility to bone metastasis [73].

2.2. PCa Affects the State and Production of PPAT

Cancer-associated adipocytes (CAA) are defined as the result of interactions between adipocytes and tumor cells, leading to the remodeling of adipocytes into a more poorly differentiated state of adipocytes, consistent with the finding that PPAT is richer in precursor adipocytes than other visceral adipose tissue [12]. This precursor cell phenotype is associated with more aggressive tumors, including PCa [52,74]. CAA is associated with the exacerbation of some of the malignant features of cancer cells, which ultimately constitute a positive feedback loop with cancer cells in the malignant progression of tumors [21,75]. For example, PCa–conditioned medium (CM) more affects preadipocytes than non-malignant PCa cells. Preadipocytes disturbed by PCa–CM participate in several biological processes of PCa progression by undergoing PCa-like tumor-like transformations [44]. Meanwhile, human PCa cells injected into the right side of thymus-free nude mice can be recruited to transplant preadipocytes on the other side. This preadipocyte migration enhances tumor growth and angiogenesis [76]. Some researchers have suggested that PCa cells can also differentiate into adipocyte-like cells and exert PPAT-like pro-cancer effects [77]. Furthermore, CAA undergoes considerable morphological and functional alterations during cancer progression. CAA undergoes dilapidation and acquires a fibroblast-like phenotype in cancer cells, especially at the front of tumor invasion [78]. This phenotypic change is often accompanied by increased secretion of adipocyte differentiation marker proteins (adiponectin, leptin and fatty acid binding proteins, and intestinal proteins) as well as the pro-inflammatory cytokines IL-6 and PAI-1 (plasminogen activator inhibitor-1). This creates a tumor microenvironment that promotes a shift in tumor cell phenotype (increased aggressiveness) [78]. For example, the expression of inflammatory factors (IL-6, IL-8, G-CSF) is significantly higher in PPAT than in serum. IL-6 expression is 375 times higher in PPAT than in serum and significantly correlates with pathological disease grade [13]. This suggests the possibility that PCa and PPAT are interrelated and is potential evidence for the evolution of PPAT into tumor-associated fibroblasts [79].
Visceral and subcutaneous adipose tissue are also associated with more aggressive pathological features of PCa. The interaction of PCa with adipose metabolism and testosterone and the effects of 5a reductase inhibitors on prostate and peripheral adipose suggest that a cross-talk may exist between periprostatic adipose tissue and tumors [80]. PCa also regulates the production of leptin via fat-resident regulatory T cells and several leptin receptors, which are associated with the quality of periglandular adipose tissue [81,82]. In vitro experiments of differentiated adipocytes and tumor cells without direct contact have also confirmed that paracrine cytokines from tumor cells induce lipolysis in adipocytes and promote the release of free fatty acids [83]. This also alters the fatty acid composition of PPAT. For example, a study compared the fatty acid composition of PPAT from 12 PCa patients and 11 patients with BPH (benign prostatic hyperplasia) confirmed high levels of palmitic acid and dihomo-γ-linolenic acid and low levels of arachidonic acid in PPAT [84]. Similarly, magnetic resonance spectroscopy showed that PPAT from patients with higher levels of PCa contained a higher ratio of monounsaturated/saturated fatty acids in the fatty acids [16,64]. Therefore, studying the specific processes by which PCa interferes with the release and composition of PPAT fatty acids may give potential avenues for intervention and treatment of PCa. In conclusion, these results suggest that PCa can regulate the state of PPAT and the release of biomolecules to accelerate PCa development in various ways.

3. Exosomes as a Mediator of Positive Feedback between Adipose Tissue and Cancer

Recent studies related to adipocytes and extracellular vesicles (EVs) of cancer origin have provided new understanding of the interactions between adipocytes and tumors. Adipose exosomes play a crucial role in cancer progression by transporting fatty acids, biooxidative enzymes, protein degrading enzymes, metabolites, and multiple non-coding RNAs into cancer cells [85,86,87,88] (Table 2). Tumor exosomes can also transport miRNAs, circRNAs, adipokines, and inflammatory factors to adipocytes, thus influencing adipose tissue differentiation and substance release. This can make the adipose further evolve into tumor-associated adipocytes, thereby providing an environment for cancer survival and progression (Table 3). Therefore, the exosomes may act as mediators for PPAT-PCa cross-talk.

3.1. Tumor Exosomes Mediate the Alteration of Adipose Tissue and Release of Biomolecules

Exosomes contain mRNA, miRNA, lncRNA, circRNA, DNA, and bioactive cellular metabolites. As a result, exosomes are involved in cellular communication and biological regulation by transporting these substances to the target receptor cells [110]. The miRNA profile of tumor-derived exosomes is similar to that of the tumor cells of origin. Dysregulated exosomal miRNAs can promote cancer migration and proliferation [111]. Tumor-derived exosomes deliver specific miRNAs (miRNA-144, miRNA-126, and miRNA-155) from breast cancer cells to peripheral adipocytes to transform resident cells. For example, exosomal miRNA-144 acts as a mediator between tumor cells and adipocytes, promoting the beige/brown differentiation of adipocytes [99,112]. Adipocytes in melanoma patients secrete more specific exosomes (containing more proteins related to fatty acid oxidation (FAO)), which induce metabolic reprogramming of tumor cells to favor FAO and promote cancer cell aggressiveness [85]. Similarly, exosomes from pancreatic cancer cells can also promote lipolysis by activating p38 and extracellular signaling regulation (ERK1/2) to phosphorylate hormone-sensitive lipases. These catabolized lipids provide energy for the survival of nearby tumor cells [109,113]. In addition, the hepatocellular carcinoma cell line HepG2 secretes exosomes containing specific proteins and activated phosphokinases to act on the NF-κB signaling pathway in adipocytes, thereby releasing adipose exosomes that promote hepatocellular carcinoma growth and angiogenesis and recruit more macrophages [114]. Cancer cell exosomes can also transport HOTAIR (HOX transcript antisense RNA) to endothelial cells, thus increasing VEGFA expression to stimulate angiogenesis, explaining why most cancer-cell-derived exosomes can promote angiogenesis [115]. Released exosomes from gastric cancer cells deliver ciRS-133 to preadipocytes, thus regulating preadipocyte differentiation to brown adipocytes by activating the PR structural domain of PRDM16 [114]. In conclusion, cancer cells differentiate adipocytes into CAA via exosomes, providing a potential material basis for their own survival and progression.

3.2. Adipose Exosomes Induce the Development of Multiple Tumors

Currently, studies on exosomes have mainly focused on cancer-cell-derived exosomes, and few studies have focused on CAA exosomes. In fact, the exosomes released from adipocytes cross-talked by tumor cells have obvious structural and functional changes [85]. For example, melanoma-associated adipocyte exosomes contain proteins involved in FAO and induce metabolic reprogramming in tumor cells. The number of secreted exosomes and their effect on tumor aggressiveness are further amplified by the dual effect of obesity and cancer, which exacerbates the symbiotic relationship between adipocytes and cancer cells [85]. For example, MMP3 is highly expressed in adipocyte-derived exosomes in obese lung cancer patients, which is usually transferred to lung cancer cells. As a result, MMP3 promotes the activity of MMP9 in lung cancer cells, thus mediating cancer cell invasion in vitro and in vivo [87]. The specific miRNAs are upregulated in CAA exosomes, inducing survival and progression of many cancer cells. For example, miR-21 is significantly upregulated in triple-negative breast and colorectal-cancer-associated adipose exosomes. Exosomes transport miR-21 from adipose tissue to cancer cells, inhibit apoptosis and promote chemoresistance by binding to apoptosis protease-activating factor-1 (APAF1) [89]. Similarly, miR-210 in exosomes released from adipose stem cells promotes endothelial cell proliferation, invasion, and migration, thus mediating tumor angiogenesis by targeting RUNX3 [116]. The miR-27a-3p in adipocyte-secreted exosomes targets ICOS, an immune-microenvironment-related gene that is significantly upregulated in obese lung adenocarcinoma (LUAD) patients, to promote antitumor immunity in LUAD [96]. In addition, exosomes released from adipose tissue in obese mice contain pro-inflammatory proteins. These exosomes can produce potential stimulatory effects in local and peripheral adipose tissue sites, thus mediating the obesity-associated inflammatory response and insulin resistance and promoting the progression of several cancers [117]. Therefore, CAA exosomes play a crucial role in tumor progression.

3.3. Exosomes Mediate Cross-Talk between PPAT and PCa

Adipose tissue distributed in the periphery of the gland does not act directly on cancer cells by releasing multiple cytokines or by creating a hypoxic environment as adipocytes in the interstitial stroma of the gland do. Studies have reported that IL-6 levels are significantly higher in PCa peripheral adipose tissue (PPAT) than in serum (100-fold). Furthermore, IL-6 levels in peripheral blood are not correlated with IL-6 levels in PPAT. Notably, IL-6 levels in PPAT are correlated with higher Gleason scores, while serum IL-6 levels are not correlated with higher Gleason scores [13]. This suggests that PPAT may not affect the level of inflammatory factors in the body and thus the progression of cancer by directly releasing inflammatory factors. Since the prostate is often separated from the surrounding PPAT by a fibromuscular capsule in the physiological state and the surrounding adipose tissue is more distant than the interstitial adipocytes of the tumor, tight cellular junctions and paracrine secretion as well as the long-distance blood transport of tumor cytokines may not be the main ways to affect it [40,118]. So, in what way does PCa mediate this process? It is certain that a more efficient mediator of crosstalk exists between PPAT and PCa. Exploring this mediator and interfering with it may be a new avenue for PCa therapy. Unlike normal stem cells, adipose stem cells cultured with PC cell-conditioned medium (CM) can form prostate-like tumor lesions in vivo and replicate invasive PCa cells in targeted tissues. This may be due to PCa cell-derived exosomes transporting oncogenic-factor-related transcripts (H-ras and K-ras), multiple miRNAs (miR-125b, miR-130b, and miR-155), and the Ras superfamily of GTPases (Rab1a, Rab1b, and Rab11a) [44]. Also, miR-145-containing exosomes released from adipose stem cells exhibit growth-inhibiting properties of PCa cells [119]. Although this contradicts the ability of PCa cell CM of inducing proliferation of PC3 cell lines and endothelial cells, it suggests that bidirectional exosomes can be released from adipose tissue disturbed by PCa cells. However, the role of adipose exosomes in PPAT with PCa is unclear. Regardless, only a few studies have reported on PCa and adipose exosomes. A systematic evaluation and meta-analysis of 86,490 patients with PCa found an association between obesity and biochemical recurrence after radical prostatectomy [120]. Obesity status of “overweight” and “obese” PCa patients is a key risk factor for biochemical recurrence after radical PCa treatment [121]. Therefore, it is crucial to investigate the interaction between PPAT and PCa, especially the role of exosomes, to provide a basis for explaining the susceptibility to biochemical recurrence after radical PCa resection in obese patients.

4. Obesity Alters the Number and Structure of Exosomes Involved in the Cross-Talk between Adiposity and Cancer

4.1. Obesity Affects the Number and Status of Adipose Exosomes

Adipocytes in obese individuals rapidly grow into mature adipocytes due to the infiltration of inflammatory factors and the construction of a hypoxic environment affecting the various stages of adipocyte differentiation [122,123,124]. Mature adipocytes secrete different populations of extracellular vesicles (EVs), including small extracellular vesicles (sEVs) and large extracellular vesicles (lEVs). The obvious intrinsic proteins of lEVs are caveolin-1, flotillin-2, and β-actin, which are involved in the shedding of microvesicles. sEVs contain endosomal sorting complex Alix, CD9, CD63, and CD81, and have high cholesterol levels [125]. The differential expression of these exosomal surface proteins suggests that they can be used in clinics [126]. During adipose immaturity, adipocytes in the undifferentiated state secrete increased levels of exosomes. These exosomes contain higher levels of arachidonic acid and adipogenic markers (PPARγ and PREF1). Interestingly, adiponectin expression in exosomes was evident at day 15 of adipose precursor cell differentiation. The uptake of these high adiponectin-expressing exosomes by tumor cells accelerates the rapid growth of various tumors, including breast cancer [43]. In vivo studies have shown that injection of adipose-derived exosomes into mice induces insulin resistance, promotes macrophage polarization processes, and stimulates the secretion of pro-inflammatory cytokines, thus promoting the release of adipose exosomes [117]. Therefore, exosome contents depend on the stage of adipocyte differentiation. Obesity can mediate exosome alterations, thus inducing tumor growth by affecting the differentiation potential of adipocytes.

4.2. Obesity Alters the Cargo and Function of Adipose Exosomes

Obesity can directly or indirectly induce cancer progression by affecting adipocyte-derived exosomal cargo. Dysfunctional adipocytes in obese subjects impair the assembly and classification of biological components in exosomes. Obesity also alters the metabolic state, thus affecting the payload and function of adipocyte exosomes. For example, a study characterized exosomes released from adipose tissue from seven obese patients (age: 12–17.5 years, BMI: 33–50 kg/m2) and five lean patients (age: 11–19 years, BMI: 22–25 kg/m2) and found that 55 miRNAs were differentially expressed in obese vs. lean visceral adipose donors (p < 0.05; fold change ≥ |1.2|). The obesity-derived exosomes can also downregulate ACVR2B and regulate the TGF-β1/Wnt/β signaling pathway in A549 cells. Exosomes released from insulin-resistant adipocytes through exposure to pro-inflammatory cytokines (highly expressed in the obese state) or adipose tissue isolated from adult subjects with T2D upregulates genes related to migration, invasiveness, and EMT. Such exosomes exhibit pro-cancer stem-cell-like cell formation and pro-invasive and pro-metastatic behaviors in breast cancer cell models compared with insulin-sensitive adipocytes or adipocytes isolated from adipose tissue of non-diabetic subjects [127]. Studies using a palmitate-induced hypertrophic 3T3-L1 adipocyte model found increased levels of miR-802-5p in the secreted exosomes of these adipocytes. In turn, miR-802-5p downregulates heat shock protein 60 (HSP60), upregulates CCAAT/enhancer binding protein (C/EBP) homologs, and enhances oxidative stress and phosphorylation of c Jun NH(2) terminal kinase (JNK) and insulin receptor substrate 1 (IRS1), leading to the development of insulin resistance [128]. The insulin-resistance-induced diabetes and exosomes with highly expressed miR-802-5p in serum are associated with tumor progression [129]. Obesity accelerates the enlargement of the fat pad, leading to hypoxia within the adipose tissue. Hypoxia upregulates adipocyte exosomal proteins associated with metabolic processes. Studies using the 3T3-L1 adipocyte model [130] showed that exosomal proteins related to lipid synthesis (acetyl-CoA carboxylase, glucose-6-phosphate dehydrogenase, and fatty acid synthase) are upregulated under hypoxic conditions (three to four times higher than under normoxic conditions) [130]. Indeed, studies in obese patients have confirmed the effect of obesity on adipocyte-derived exosomal cargo. For example, clinical studies have found that subcutaneous adipocyte-derived exosomes from obese patients contain higher levels of proteins associated with FAO [85]. In addition, exosomes released from large adipocytes can be transferred to small adipocytes, thereby stimulating adipogenesis and hypertrophy of small adipocytes [131]. These exosomes may be transported into the tumor mesenchyme through the bloodstream, inducing maturation and differentiation of cancer-associated adipocytes and thus providing sufficient energy and material for cancer progression.

4.3. Adipose Exosomes Mediate PCa Progression in Obese Patients

Current research on the causes of obesity-mediated cancer progression focuses on soluble factors, such as increased circulating levels of insulin and other growth factors, altered inflammatory status, and the release of inflammatory molecules and fatty acids from dyslipidemia [8,132]. However, obesity also increases the migration rate of preadipocytes in WAT, thereby mediating PCa progression [133]. In the case of PPAT, obesity also alters the gene expression profile of PPAT, promoting proliferation and immune escape of cancer cells. This creates a microenvironment conducive to PCa progression [134]. Obese men have higher levels of angiogenic capacity in PPAT than lean individuals [71]. Several studies have also indicated that adipocytes can directly construct the tumor microenvironment, forming an adipose tissue containing multiple cells, including immune cells, fibroblasts, preadipose stem cell populations, and mature adipocytes. These cells are associated with the formation and development of various cancers [135,136]. Furthermore, increasing evidence has shown that adipocyte EVs have a key role in tumor progression and tumor-cell–adipocyte interactions in obese patients [137,138]. An experiment investigating the effect of 3T3-L1-derived EVs on the invasiveness of PC3 and DU145 cells found that EVs released from adipocytes stimulate the proliferation of PCa cells and significantly enhance their migratory and invasive abilities. This could be because adipose EVs can trigger a rapid glycolytic process, thus increasing glucose uptake, lactate release, and ATP synthesis in PCa cells [37]. EVs also causes AKT activation and HIF-1α stabilization, commonly associated with overproliferation, metastasis, and chemoresistance in PCa [139,140,141,142]. This recent study suggests that exosomes released in the obese state may be important in the onset and progression of PCa. Taken together, these studies bring new insights into the impact of under-studied adipose deposits, especially PPAT, and their released EVs on PCa (Figure 2).

5. Conclusions

The biological processes in adipose tissue, which are cancer-associated, are constantly disturbed by cancer, either directly or indirectly, and this in turn accelerates the further spread of cancer cells in some possible ways [50,51,84]. Especially for the peripheral tissues of cancerous tissues, this symbiosis suggests that cancer may not only communicate with cells in the vicinity of the tumor tissue mesenchyme but may also have some crosstalk with tissues relatively distant from the cancerous tissue, such as peripheral adipose. Therefore, it is important to explore the mechanism of this mutual cross-talk to confirm the existence of this relationship and explain obesity as a cancer risk factor. Exosomes are biological vesicles secreted by cells and can mediate cancer progression by transporting various oncogenic molecules [143]. Exosomes, as biological vesicles secreted by cells with substance transport, signaling, and low antigenic and easy circulation characteristics [144], can mediate cancer progression by transporting various oncogenic molecules [145,146]. Also, tumor-derived exosomes can be transported to adipose tissue to participate in adipocyte remodeling [85,146]. These results suggest that exosomes play a key role in the mutual cross-talk between peripheral adiposity and cancer. Obesity exacerbates this process and can directly alter the release of multiple cytokines from adipocytes, thus influencing cancer development and progression. Obesity can also indirectly promote tumor survival and progression by inducing related diseases [133]. Obesity also exacerbates the positive feedback between cancer and adipose tissue by interfering with the production and cargo of adipose exosomes and thus remotely controlling the cross-talk between adiposity and cancer [37]. Therefore, the effects of obesity and adipose exosomes on PCa and PPAT during their mutual crosstalk are bound to be the focus of future studies.

6. Discussion

Obesity is a risk factor for many malignancies, including PCa, colon, breast, liver, and pancreatic cancers. The induction of cytokine release, vascular microenvironment formation, and tumor mesenchymal cell remodeling are all common pathways by which obesity mediates cancer progression. Indeed, the complexity of cancer progression suggests that tumor cells can act not only directly on themselves to maintain their own survival but also indirectly by altering the tumor microenvironment including the release of specific cytokines or vesicles to maintain tumor cell survival or progression. In addition, there appears to be a more powerful disturbing effect of tumor cells on peritumor tissues. For example, tumor tissue can not only affect interstitial cells but can also further induce cancer development by potentially interfering with the state of the tissue surrounding the cancer [37,50,147]. However, there are no robust studies on how tumor cells interfere with surrounding tissues and how surrounding tissues mediate the progression of cancer. Similarly, there is still a lack of sufficient evidence on how peri-cancerous fat is disturbed by cancer and how peri-cancerous fat further contributes to cancer progression.
Whether the presence of PPAT as one of the possible factors of PCa progression may influence the further spread of the lesion leading to multiple recurrences of the cancer as well as maintaining high carcinogenicity and carcinogenicity in the body and thus affecting the use of chemotherapeutic drugs, etc. is uncertain. This leads one to consider the need for simultaneous excision of periglandular adipose tissue during radical surgery for PCa. PPAT is more distant than tumor mesenchymal adipocytes. Therefore, they may not interact as mesenchymal cells interact, such as tight junctions and paracrine secretion. Unlike adipose tissues in other parts of the body, inflammatory factors are highly expressed in PPAT [13]. This suggests the existence of a new mediator that is mediating the mutual crosstalk between PCa and PPAT. Here, we propose that exosomes may be an important way to mediate this process. For example, several studies have shown that exosomes released from PCa cells can mediate stem cell remodeling of adipose tissue [145] and adipose stem-cell-derived exosomes mediate the inhibition of PCa growth via PCa stromal cells [119]. In addition, interfering with the production of adipose exosomes may be an effective way to halt the progression of PCa and to inhibit the crosstalk between PPAT and PCa. The drugs that inhibit vesicle production and release and affect the expression of exosome surface membrane proteins can also reduce exosome production. However, these drugs still have deadly side effects, and more research is needed to confirm their exact value in oncology treatment. Many cancer cells, including PCa can release both pro- and anti-cancer bipartite exosomes, and adipocytes can also release exosomes that activate inflammatory responses in the tumor microenvironment and thus inhibit cancer proliferation and spread [148,149,150]. Therefore, the extraction of potential oncogenic exosomes from adipose exosomes may promote tumor targeting therapy. However, isolating and identifying these specific exosomes is difficult, thus limiting the exosome research work. Adipose exosome research has widely employed the method of interfering with the expression of specific oncogenic molecules in exosomes. Drugs interfering with the source cells or exosome delivery disruption tools can effectively inhibit PCa progression.
Exosome therapies overcome the challenges associated with cellular therapies, including immune rejection, tumorigenic controversy, tissue-specific antigen targeting on the membrane surface, low immunogenicity, and non-invasive passage through the tissue barrier. In addition, adipose tissue may be potentially more promising for use than other tissue exosomes, as follows. (1). Tumor-derived exosomes can activate macrophages in adipose tissue. More monocytes/macrophages may be recruited to adipose tissue after the activation of resident macrophages, further enhancing the inflammatory response. This inflammatory environment further promotes the proliferation of adipocytes and increases the number of associated exosomes. The increased adipocyte size caused by obesity may result in the release of more exosomes from adipose tissue. The combined effect is more efficient for exosome acquisition. (2). The ease of access to adipose tissue and the simplicity of ethical review (Figure 3). Furthermore, liposuction, the most common plastic surgery procedure in the 20th century, can obtain large amounts of human-derived adipose tissue [151]. This access to large amounts of adipose tissue offers the possibility of bulk acquisition and use of human-derived exosomes. (3). Current research on exosomes focuses on MSC exosomes, especially on the regenerative repair functions of MSC exosomes, such as improvement of recalcitrant hair loss, post-traumatic skin healing, spinal cord injury repair, and osteoarthritis [119,152,153]. A prospective, faceted, randomized placebo-controlled trial confirmed that adipose MSC exosomes can be used in skin brightening. The preparations containing ASC exosomes significantly reduce melanin levels than placebo [154]. These studies indicate that the use of MSC exosomes is very promising, and the production of MSC exosomes is bound to become a major concern, and adipose tissue, as the most important source of known MSCs, is bound to become a priority for future sources of large-scale production of stem cell exosomes.

Author Contributions

G.Z. conceived the manuscript, S.F. searched publications and draft the manuscript. K.L. and C.L. edited tables and figures. X.Z. and J.Z. reviewed the manuscript and polished the grammar. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (grant nos. 81760462 and 81860456). This research was supported by Science and Technology Research Project of Jiangxi Provincial Education Department (grant nos. GJJ180789). This study was also supported by Leading Talents Program of Jiangxi Provincial Major Discipline Academic and Technical Leaders Training Program (grant nos. 20213BCJL22038).

Acknowledgments

All figures are created with BioRender.com (accessed on 18 August 2022).

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Li, J.; Xu, C.; Lee, H.J.; Ren, S.; Zi, X.; Zhang, Z.; Wang, H.; Yu, Y.; Yang, C.; Gao, X.; et al. A genomic and epigenomic atlas of prostate cancer in Asian populations. Nature 2020, 580, 93–99. [Google Scholar] [CrossRef]
  3. Zhong, S.; Yan, X.; Wu, Y.; Zhang, X.; Chen, L.; Tang, J.; Zhao, J. Body mass index and mortality in prostate cancer patients: A dose-response meta-analysis. Prostate Cancer Prostatic Dis. 2016, 19, 122–131. [Google Scholar] [CrossRef] [PubMed]
  4. Hu, M.B.; Liu, S.H.; Jiang, H.W.; Bai, P.D.; Ding, Q. Obesity affects the biopsy-mediated detection of prostate cancer, particularly high-grade prostate cancer: A dose-response meta-analysis of 29,464 patients. PLoS ONE 2014, 9, e106677. [Google Scholar] [CrossRef] [PubMed]
  5. Allott, E.H.; Howard, L.E.; Song, H.J.; Sourbeer, K.N.; Koontz, B.F.; Salama, J.K.; Freedland, S.J. Racial differences in adipose tissue distribution and risk of aggressive prostate cancer among men undergoing radiotherapy. Cancer Epidemiol. Biomark. Prev. 2014, 23, 2404–2412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Bhindi, B.; Trottier, G.; Elharram, M.; Fernandes, K.A.; Lockwood, G.; Toi, A.; Hersey, K.M.; Finelli, A.; Evans, A.; van der Kwast, T.H.; et al. Measurement of peri-prostatic fat thickness using transrectal ultrasonography (TRUS): A new risk factor for prostate cancer. BJU Int. 2012, 110, 980–986. [Google Scholar] [CrossRef]
  7. Zadra, G.; Photopoulos, C.; Loda, M. The fat side of prostate cancer. Biochim. Et Biophys. Acta 2013, 1831, 1518–1532. [Google Scholar] [CrossRef] [Green Version]
  8. Grossmann, M.; Wittert, G. Androgens, diabetes and prostate cancer. Endocr.-Relat. Cancer 2012, 19, F47–F62. [Google Scholar] [CrossRef] [Green Version]
  9. Billis, A. Intraprostatic fat: Does it exist? Hum. Pathol. 2004, 35, 525. [Google Scholar] [CrossRef]
  10. van Roermund, J.G.; Bol, G.H.; Witjes, J.A.; Ruud Bosch, J.L.; Kiemeney, L.A.; van Vulpen, M. Periprostatic fat measured on computed tomography as a marker for prostate cancer aggressiveness. World J. Urol. 2010, 28, 699–704. [Google Scholar] [CrossRef]
  11. Salji, M.; Hendry, J.; Patel, A.; Ahmad, I.; Nixon, C.; Leung, H.Y. Peri-prostatic Fat Volume Measurement as a Predictive Tool for Castration Resistance in Advanced Prostate Cancer. Eur. Urol. Focus 2018, 4, 858–866. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Ribeiro, R.; Monteiro, C.; Silvestre, R.; Castela, A.; Coutinho, H.; Fraga, A.; Príncipe, P.; Lobato, C.; Costa, C.; Cordeiro-da-Silva, A.; et al. Human periprostatic white adipose tissue is rich in stromal progenitor cells and a potential source of prostate tumor stroma. Exp. Biol. Med. Maywood 2012, 237, 1155–1162. [Google Scholar] [CrossRef] [PubMed]
  13. Finley, D.S.; Calvert, V.S.; Inokuchi, J.; Lau, A.; Narula, N.; Petricoin, E.F.; Zaldivar, F.; Santos, R.; Tyson, D.R.; Ornstein, D.K. Periprostatic adipose tissue as a modulator of prostate cancer aggressiveness. J. Urol 2009, 182, 1621–1627. [Google Scholar] [CrossRef] [PubMed]
  14. van Roermund, J.G.; Hinnen, K.A.; Tolman, C.J.; Bol, G.H.; Witjes, J.A.; Bosch, J.L.; Kiemeney, L.A.; van Vulpen, M. Periprostatic fat correlates with tumour aggressiveness in prostate cancer patients. BJU Int. 2011, 107, 1775–1779. [Google Scholar] [CrossRef] [PubMed]
  15. Zhang, Q.; Sun, L.J.; Qi, J.; Yang, Z.G.; Huang, T.; Huo, R.C. Periprostatic adiposity measured on magnetic resonance imaging correlates with prostate cancer aggressiveness. Urol. J. 2014, 11, 1793–1799. [Google Scholar]
  16. Woo, S.; Cho, J.Y.; Kim, S.Y.; Kim, S.H. Periprostatic fat thickness on MRI: Correlation with Gleason score in prostate cancer. AJR Am. J. Roentgenol. 2015, 204, W43–W47. [Google Scholar] [CrossRef]
  17. Cao, Y.; Cao, M.; Chen, Y.; Yu, W.; Fan, Y.; Liu, Q.; Gao, G.; Zhao, Z.; Wang, X.; Jin, J. The combination of prostate imaging reporting and data system version 2 (PI-RADS v2) and periprostatic fat thickness on multi-parametric MRI to predict the presence of prostate cancer. Oncotarget 2017, 8, 44040–44049. [Google Scholar] [CrossRef]
  18. Dahran, N.; Szewczyk-Bieda, M.; Wei, C.; Vinnicombe, S.; Nabi, G. Normalized periprostatic fat MRI measurements can predict prostate cancer aggressiveness in men undergoing radical prostatectomy for clinically localised disease. Sci. Rep. 2017, 7, 4630. [Google Scholar]
  19. Gregg, J.R.; Surasi, D.S.; Childs, A.; Moll, N.; Ward, J.F.; Kim, J.; Daniel, C.R.; Logothetis, C.; Bathala, T.; Davis, J.W. The Association of Periprostatic Fat and Grade Group Progression in Men with Localized Prostate Cancer on Active Surveillance. J Urol. 2021, 205, 122–128. [Google Scholar] [CrossRef]
  20. Sasaki, T.; Sugino, Y.; Kato, M.; Nishikawa, K.; Kanda, H. Pre-treatment ratio of periprostatic to subcutaneous fat thickness on MRI is an independent survival predictor in hormone-naïve men with advanced prostate cancer. Int. J. Clin. Oncol. 2020, 25, 370–376. [Google Scholar] [CrossRef]
  21. Ribeiro, R.; Monteiro, C.; Cunha, V.; Oliveira, M.J.; Freitas, M.; Fraga, A.; Príncipe, P.; Lobato, C.; Lobo, F.; Morais, A.; et al. Human periprostatic adipose tissue promotes prostate cancer aggressiveness in vitro. J. Exp. Clin. Cancer Res. 2012, 31, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. La Civita, E.; Liotti, A.; Cennamo, M.; Crocetto, F.; Ferro, M.; Liguoro, P.; Cimmino, A.; Imbimbo, C.; Beguinot, F.; Formisano, P.; et al. Peri-Prostatic Adipocyte-Released TGFβ Enhances Prostate Cancer Cell Motility by Upregulation of Connective Tissue Growth Factor. Biomedicines 2021, 9, 1692. [Google Scholar] [CrossRef] [PubMed]
  23. Figiel, S.; Pinault, M.; Domingo, I.; Guimaraes, C.; Guibon, R.; Besson, P.; Tavernier, E.; Blanchet, P.; Multigner, L.; Bruyère, F.; et al. Fatty acid profile in peri-prostatic adipose tissue and prostate cancer aggressiveness in African-Caribbean and Caucasian patients. Eur. J. Cancer Oxf. Engl. 1990 2018, 91, 107–115. [Google Scholar] [CrossRef]
  24. Liotti, A.; La Civita, E.; Cennamo, M.; Crocetto, F.; Ferro, M.; Guadagno, E.; Insabato, L.; Imbimbo, C.; Palmieri, A.; Mirone, V.; et al. Periprostatic adipose tissue promotes prostate cancer resistance to docetaxel by paracrine IGF-1 upregulation of TUBB2B beta-tubulin isoform. Prostate 2021, 81, 407–417. [Google Scholar] [CrossRef] [PubMed]
  25. Laurent, V.; Toulet, A.; Attané, C.; Milhas, D.; Dauvillier, S.; Zaidi, F.; Clement, E.; Cinato, M.; Le Gonidec, S.; Guérard, A.; et al. Periprostatic Adipose Tissue Favors Prostate Cancer Cell Invasion in an Obesity-Dependent Manner: Role of Oxidative Stress. Mol. Cancer Res. MCR 2019, 17, 821–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Xu, L.; Shen, M.; Chen, X.; Zhu, R.; Yang, D.R.; Tsai, Y.; Keng, P.C.; Chen, Y.; Lee, S.O. Adipocytes affect castration-resistant prostate cancer cells to develop the resistance to cytotoxic action of NK cells with alterations of PD-L1/NKG2D ligand levels in tumor cells. Prostate 2018, 78, 353–364. [Google Scholar] [CrossRef]
  27. Fontaine, A.; Bellanger, D.; Guibon, R.; Bruyère, F.; Brisson, L.; Fromont, G. Lipophagy and prostate cancer: Association with disease aggressiveness and proximity to periprostatic adipose tissue. J. Pathol. 2021, 255, 166–176. [Google Scholar] [CrossRef]
  28. Sacca, P.A.; Creydt, V.P.; Choi, H.; Mazza, O.N.; Fletcher, S.J.; Vallone, V.B.; Scorticati, C.; Chasseing, N.A.; Calvo, J.C. Human periprostatic adipose tissue: Its influence on prostate cancer cells. Cell Physiol Biochem. 2012, 30, 113–122. [Google Scholar] [CrossRef]
  29. Avgerinos, K.I.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metab. Clin. Exp. 2019, 92, 121–135. [Google Scholar] [CrossRef]
  30. Kelly, S.P.; Graubard, B.I.; Andreotti, G.; Younes, N.; Cleary, S.D.; Cook, M.B. Prediagnostic Body Mass Index Trajectories in Relation to Prostate Cancer Incidence and Mortality in the PLCO Cancer Screening Trial. J. Natl. Cancer Inst. 2017, 109, djw225. [Google Scholar] [CrossRef] [Green Version]
  31. Pernar, C.H.; Ebot, E.M.; Wilson, K.M.; Mucci, L.A. The Epidemiology of Prostate Cancer. Cold Spring Harb. Perspect. Med. 2018, 8, a030361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. De Nunzio, C.; Freedland, S.J.; Miano, L.; Finazzi Agrò, E.; Bañez, L.; Tubaro, A. The uncertain relationship between obesity and prostate cancer: An Italian biopsy cohort analysis. Eur. J. Surg. Oncol. 2011, 37, 1025–1029. [Google Scholar] [CrossRef] [PubMed]
  33. Møller, H.; Roswall, N.; Van Hemelrijck, M.; Larsen, S.B.; Cuzick, J.; Holmberg, L.; Overvad, K.; Tjønneland, A. Prostate cancer incidence, clinical stage and survival in relation to obesity: A prospective cohort study in Denmark. Int. J. Cancer 2015, 136, 1940–1947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Guh, D.P.; Zhang, W.; Bansback, N.; Amarsi, Z.; Birmingham, C.L.; Anis, A.H. The incidence of co-morbidities related to obesity and overweight: A systematic review and meta-analysis. BMC Public Health 2009, 9, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Strong, A.L.; Burow, M.E.; Gimble, J.M.; Bunnell, B.A. Concise review: The obesity cancer paradigm: Exploration of the interactions and crosstalk with adipose stem cells. Stem Cells Dayt. Ohio 2015, 33, 318–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kwan, H.Y.; Chen, M.; Xu, K.; Chen, B. The impact of obesity on adipocyte-derived extracellular vesicles. Cell. Mol. Life Sci. CMLS 2021, 78, 7275–7288. [Google Scholar] [CrossRef]
  37. Fontana, F.; Anselmi, M.; Carollo, E.; Sartori, P.; Procacci, P.; Carter, D.; Limonta, P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022, 11, 2388. [Google Scholar] [CrossRef]
  38. Kubo, H. Extracellular Vesicles in Lung Disease. Chest 2018, 153, 210–216. [Google Scholar] [CrossRef]
  39. Couzin, J. Cell biology: The ins and outs of exosomes. Science 2005, 308, 1862–1863. [Google Scholar] [CrossRef]
  40. Hong, H.; Koch, M.O.; Foster, R.S.; Bihrle, R.; Gardner, T.A.; Fyffe, J.; Ulbright, T.M.; Eble, J.N.; Cheng, L. Anatomic distribution of periprostatic adipose tissue: A mapping study of 100 radical prostatectomy specimens. Cancer 2003, 97, 1639–1643. [Google Scholar] [CrossRef]
  41. Byar, D.P.; Mostofi, F.K. Carcinoma of the prostate: Prognostic evaluation of certain pathologic features in 208 radical prostatectomies. Examined by the step-section technique. Cancer 1972, 30, 5–13. [Google Scholar] [CrossRef]
  42. Cheng, L.; Darson, M.F.; Bergstralh, E.J.; Slezak, J.; Myers, R.P.; Bostwick, D.G. Correlation of margin status and extraprostatic extension with progression of prostate carcinoma. Cancer 1999, 86, 1775–1782. [Google Scholar] [CrossRef]
  43. Nieman, K.M.; Kenny, H.A.; Penicka, C.V.; Ladanyi, A.; Buell-Gutbrod, R.; Zillhardt, M.R.; Romero, I.L.; Carey, M.S.; Mills, G.B.; Hotamisligil, G.S.; et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nat. Med. 2011, 17, 1498–1503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Abd Elmageed, Z.Y.; Yang, Y.; Thomas, R.; Ranjan, M.; Mondal, D.; Moroz, K.; Fang, Z.; Rezk, B.M.; Moparty, K.; Sikka, S.C.; et al. Neoplastic reprogramming of patient-derived adipose stem cells by prostate cancer cell-associated exosomes. Stem. Cells 2014, 32, 983–997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sakers, A.; De Siqueira, M.K.; Seale, P.; Villanueva, C.J. Adipose-tissue plasticity in health and disease. Cell 2022, 185, 419–446. [Google Scholar] [CrossRef]
  46. Moreira, Â.; Pereira, S.S.; Costa, M.; Morais, T.; Pinto, A.; Fernandes, R.; Monteiro, M.P. Adipocyte secreted factors enhance aggressiveness of prostate carcinoma cells. PLoS ONE 2015, 10, e0123217. [Google Scholar] [CrossRef] [Green Version]
  47. Onuma, M.; Bub, J.D.; Rummel, T.L.; Iwamoto, Y. Prostate cancer cell-adipocyte interaction: Leptin mediates androgen-independent prostate cancer cell proliferation through c-Jun NH2-terminal kinase. J. Biol. Chem. 2003, 278, 42660–42667. [Google Scholar] [CrossRef] [Green Version]
  48. Ito, Y.; Ishiguro, H.; Kobayashi, N.; Hasumi, H.; Watanabe, M.; Yao, M.; Uemura, H. Adipocyte-derived monocyte chemotactic protein-1 (MCP-1) promotes prostate cancer progression through the induction of MMP-2 activity. Prostate 2015, 75, 1009–1019. [Google Scholar] [CrossRef]
  49. Pettersson, A.; Lis, R.T.; Meisner, A.; Flavin, R.; Stack, E.C.; Fiorentino, M.; Finn, S.; Graff, R.E.; Penney, K.L.; Rider, J.R.; et al. Modification of the association between obesity and lethal prostate cancer by TMPRSS2:ERG. J. Natl. Cancer Inst. 2013, 105, 1881–1890. [Google Scholar] [CrossRef] [Green Version]
  50. Allott, E.H.; Masko, E.M.; Freedland, S.J. Obesity and prostate cancer: Weighing the evidence. Eur. Urol. 2013, 63, 800–809. [Google Scholar] [CrossRef] [Green Version]
  51. Uzunlulu, M.; Telci Caklili, O.; Oguz, A. Association between Metabolic Syndrome and Cancer. Ann. Nutr. Metab. 2016, 68, 173–179. [Google Scholar] [CrossRef] [PubMed]
  52. Laurent, V.; Guérard, A.; Mazerolles, C.; Le Gonidec, S.; Toulet, A.; Nieto, L.; Zaidi, F.; Majed, B.; Garandeau, D.; Socrier, Y.; et al. Periprostatic adipocytes act as a driving force for prostate cancer progression in obesity. Nat. Commun. 2016, 7, 10230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Duffy, M.J.; Maguire, T.M.; Hill, A.; McDermott, E.; O’Higgins, N. Metalloproteinases: Role in breast carcinogenesis, invasion and metastasis. Breast Cancer Res. BCR 2000, 2, 252–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Tokuda, Y.; Satoh, Y.; Fujiyama, C.; Toda, S.; Sugihara, H.; Masaki, Z. Prostate cancer cell growth is modulated by adipocyte-cancer cell interaction. BJU Int. 2003, 91, 716–720. [Google Scholar] [CrossRef] [Green Version]
  55. Kaneko, A.; Satoh, Y.; Tokuda, Y.; Fujiyama, C.; Udo, K.; Uozumi, J. Effects of adipocytes on the proliferation and differentiation of prostate cancer cells in a 3-D culture model. Int. J. Urol. Off. J. Jpn. Urol. Assoc. 2010, 17, 369–376. [Google Scholar] [CrossRef]
  56. Divella, R.; De Luca, R.; Abbate, I.; Naglieri, E.; Daniele, A. Obesity and cancer: The role of adipose tissue and adipo-cytokines-induced chronic inflammation. J. Cancer 2016, 7, 2346–2359. [Google Scholar] [CrossRef] [Green Version]
  57. Weisberg, S.P.; McCann, D.; Desai, M.; Rosenbaum, M.; Leibel, R.L.; Ferrante, A.W., Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 2003, 112, 1796–1808. [Google Scholar] [CrossRef]
  58. Waugh, D.J.; Wilson, C. The interleukin-8 pathway in cancer. Clin. Cancer Res. 2008, 14, 6735–6741. [Google Scholar] [CrossRef] [Green Version]
  59. Sivashanmugam, P.; Tang, L.; Daaka, Y. Interleukin 6 mediates the lysophosphatidic acid-regulated cross-talk between stromal and epithelial prostate cancer cells. J. Biol. Chem. 2004, 279, 21154–21159. [Google Scholar] [CrossRef] [Green Version]
  60. Xu, H.; Hu, M.B.; Bai, P.D.; Zhu, W.H.; Liu, S.H.; Hou, J.Y.; Xiong, Z.Q.; Ding, Q.; Jiang, H.W. Proinflammatory cytokines in prostate cancer development and progression promoted by high-fat diet. BioMed Res. Int. 2015, 2015, 249741. [Google Scholar] [CrossRef] [Green Version]
  61. Gucalp, A.; Iyengar, N.M.; Zhou, X.K.; Giri, D.D.; Falcone, D.J.; Wang, H.; Williams, S.; Krasne, M.D.; Yaghnam, I.; Kunzel, B.; et al. Periprostatic adipose inflammation is associated with high-grade prostate cancer. Prostate Cancer Prostatic Dis. 2017, 20, 418–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Novakovic, K.; Fitchev, P.; Brendler, C.; Plunkett, B.; Dangle, P.; McGuire, M.; Shevrin, D.; Cornwell, M.; Quinn, M.; Kaul, K. 314 Active triacylglycerol metabolism and increased infiltrating adipocyte density in high grade prostate cancer. J. Urol. 2012, 187, e127. [Google Scholar] [CrossRef]
  63. Gazi, E.; Gardner, P.; Lockyer, N.P.; Hart, C.A.; Brown, M.D.; Clarke, N.W. Direct evidence of lipid translocation between adipocytes and prostate cancer cells with imaging FTIR microspectroscopy. J. Lipid Res. 2007, 48, 1846–1856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Iordanescu, G.; Brendler, C.; Crawford, S.E.; Wyrwicz, A.M.; Venkatasubramanian, P.N.; Doll, J.A. MRS measured fatty acid composition of periprostatic adipose tissue correlates with pathological measures of prostate cancer aggressiveness. J. Magn. Reson. Imaging JMRI 2015, 42, 651–657. [Google Scholar] [CrossRef] [PubMed]
  65. Nieman, K.M.; Romero, I.L.; Van Houten, B.; Lengyel, E. Adipose tissue and adipocytes support tumorigenesis and metastasis. Biochim. Et Biophys. Acta 2013, 1831, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  66. Susa, T.; Ikaga, R.; Kajitani, T.; Iizuka, M.; Okinaga, H.; Tamamori-Adachi, M.; Okazaki, T. Wild-type and specific mutant androgen receptor mediates transcription via 17β-estradiol in sex hormone-sensitive cancer cells. J. Cell. Physiol. 2015, 230, 1594–1606. [Google Scholar] [CrossRef]
  67. Capurso, C.; Capurso, A. From excess adiposity to insulin resistance: The role of free fatty acids. Vasc. Pharmacol. 2012, 57, 91–97. [Google Scholar] [CrossRef]
  68. de Ferranti, S.; Mozaffarian, D. The perfect storm: Obesity, adipocyte dysfunction, and metabolic consequences. Clin. Chem. 2008, 54, 945–955. [Google Scholar] [CrossRef] [Green Version]
  69. Zhang, Y.; Daquinag, A.; Traktuev, D.O.; Amaya-Manzanares, F.; Simmons, P.J.; March, K.L.; Pasqualini, R.; Arap, W.; Kolonin, M.G. White adipose tissue cells are recruited by experimental tumors and promote cancer progression in mouse models. Cancer Res. 2009, 69, 5259–5266. [Google Scholar] [CrossRef] [Green Version]
  70. Klopp, A.H.; Zhang, Y.; Solley, T.; Amaya-Manzanares, F.; Marini, F.; Andreeff, M.; Debeb, B.; Woodward, W.; Schmandt, R.; Broaddus, R.; et al. Omental adipose tissue-derived stromal cells promote vascularization and growth of endometrial tumors. Clin. Cancer Res. 2012, 18, 771–782. [Google Scholar] [CrossRef] [Green Version]
  71. Venkatasubramanian, P.N.; Crawford, S.E.; Fitchev, P.S.; Brendler, C.B.; Plunkett, B.A.; Abroe, B.; Mafi, M.; Morgan, G.; Cornwell, M.L.; O’Leary, J. Periprostatic fat from obese patients promotes prostate cancer growth. Cancer Res. 2012, 72, 1499. [Google Scholar] [CrossRef]
  72. Zhang, T.; Tseng, C.; Zhang, Y.; Sirin, O.; Corn, P.G.; Li-Ning-Tapia, E.M.; Troncoso, P.; Davis, J.; Pettaway, C.; Ward, J.; et al. CXCL1 mediates obesity-associated adipose stromal cell trafficking and function in the tumour microenvironment. Nat. Commun. 2016, 7, 11674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Adekoya, T.O.; Richardson, R.M. Cytokines and Chemokines as Mediators of Prostate Cancer Metastasis. Int. J. Mol. Sci. 2020, 21, 4449. [Google Scholar] [CrossRef] [PubMed]
  74. Dirat, B.; Bochet, L.; Dabek, M.; Daviaud, D.; Dauvillier, S.; Majed, B.; Wang, Y.Y.; Meulle, A.; Salles, B.; Le Gonidec, S.; et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011, 71, 2455–2465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Ribeiro, R.J.; Monteiro, C.P.; Cunha, V.F.; Azevedo, A.S.; Oliveira, M.J.; Monteiro, R.; Fraga, A.M.; Príncipe, P.; Lobato, C.; Lobo, F.; et al. Tumor cell-educated periprostatic adipose tissue acquires an aggressive cancer-promoting secretory profile. Cell. Physiol. Biochem. 2012, 29, 233–240. [Google Scholar] [CrossRef]
  76. Lin, G.; Yang, R.; Banie, L.; Wang, G.; Ning, H.; Li, L.C.; Lue, T.F.; Lin, C.S. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate 2010, 70, 1066–1073. [Google Scholar] [CrossRef] [Green Version]
  77. Zhau, H.E.; He, H.; Wang, C.Y.; Zayzafoon, M.; Morrissey, C.; Vessella, R.L.; Marshall, F.F.; Chung, L.W.; Wang, R. Human prostate cancer harbors the stem cell properties of bone marrow mesenchymal stem cells. Clin. Cancer Res. 2011, 17, 2159–2169. [Google Scholar] [CrossRef] [Green Version]
  78. Bochet, L.; Lehuédé, C.; Dauvillier, S.; Wang, Y.Y.; Dirat, B.; Laurent, V.; Dray, C.; Guiet, R.; Maridonneau-Parini, I.; Le Gonidec, S.; et al. Adipocyte-derived fibroblasts promote tumor progression and contribute to the desmoplastic reaction in breast cancer. Cancer Res. 2013, 73, 5657–5668. [Google Scholar]
  79. Biffi, G.; Tuveson, D.A. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol. Rev. 2021, 101, 147–176. [Google Scholar] [CrossRef]
  80. Taussky, D.; Barkati, M.; Campeau, S.; Zerouali, K.; Nadiri, A.; Saad, F.; Delouya, G. Changes in periprostatic adipose tissue induced by 5α-reductase inhibitors. Andrology 2017, 5, 511–515. [Google Scholar]
  81. De Rosa, V.; Procaccini, C.; Calì, G.; Pirozzi, G.; Fontana, S.; Zappacosta, S.; La Cava, A.; Matarese, G. A key role of leptin in the control of regulatory T cell proliferation. Immunity 2007, 26, 241–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Ghantous, C.M.; Azrak, Z.; Hanache, S.; Abou-Kheir, W.; Zeidan, A. Differential Role of Leptin and Adiponectin in Cardiovascular System. Int. J. Endocrinol. 2015, 2015, 534320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Tisdale, M.J. Cachexia in cancer patients. Nat. Rev. Cancer 2002, 2, 862–871. [Google Scholar] [CrossRef] [PubMed]
  84. Careaga Quiroga, V.P.; Sacca, P.A.; Mazza, O.N.; Scorticati, C.; Vitagliano, G.; Fletcher, S.J.; Maier, M.S.; Calvo, J.C. Fatty acid composition of human periprostatic adipose tissue from argentine patients and its relationship to prostate cancer and benign prostatic hyperplasia. Res. Cancer Tumor 2015, 4, 4,1–6. [Google Scholar]
  85. Lazar, I.; Clement, E.; Dauvillier, S.; Milhas, D.; Ducoux-Petit, M.; LeGonidec, S.; Moro, C.; Soldan, V.; Dalle, S.; Balor, S.; et al. Adipocyte Exosomes Promote Melanoma Aggressiveness through Fatty Acid Oxidation: A Novel Mechanism Linking Obesity and Cancer. Cancer Res. 2016, 76, 4051–4057. [Google Scholar] [CrossRef]
  86. Clement, E.; Lazar, I.; Attané, C.; Carrié, L.; Dauvillier, S.; Ducoux-Petit, M.; Esteve, D.; Menneteau, T.; Moutahir, M.; Le Gonidec, S.; et al. Adipocyte extracellular vesicles carry enzymes and fatty acids that stimulate mitochondrial metabolism and remodeling in tumor cells. EMBO J. 2020, 39, e102525. [Google Scholar] [CrossRef]
  87. Wang, J.; Wu, Y.; Guo, J.; Fei, X.; Yu, L.; Ma, S. Adipocyte-derived exosomes promote lung cancer metastasis by increasing MMP9 activity via transferring MMP3 to lung cancer cells. Oncotarget 2017, 8, 81880–81891. [Google Scholar] [CrossRef] [Green Version]
  88. Liu, Y.; Tan, J.; Ou, S.; Chen, J.; Chen, L. Adipose-derived exosomes deliver miR-23a/b to regulate tumor growth in hepatocellular cancer by targeting the VHL/HIF axis. J. Physiol. Biochem. 2019, 75, 391–401. [Google Scholar] [CrossRef]
  89. Au Yeung, C.L.; Co, N.N.; Tsuruga, T.; Yeung, T.L.; Kwan, S.Y.; Leung, C.S.; Li, Y.; Lu, E.S.; Kwan, K.; Wong, K.K.; et al. Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun 2016, 7, 11150. [Google Scholar] [CrossRef] [Green Version]
  90. La Camera, G.; Gelsomino, L.; Malivindi, R.; Barone, I.; Panza, S.; De Rose, D.; Giordano, F.; D’Esposito, V.; Formisano, P.; Bonofiglio, D.; et al. Adipocyte-derived extracellular vesicles promote breast cancer cell malignancy through HIF-1α activity. Cancer Lett. 2021, 521, 155–168. [Google Scholar] [CrossRef]
  91. Guo, G.; Tan, Z.; Liu, Y.; Shi, F.; She, J. The therapeutic potential of stem cell-derived exosomes in the ulcerative colitis and colorectal cancer. Stem Cell Res. Ther. 2022, 13, 138. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, H.; Deng, T.; Ge, S.; Liu, Y.; Bai, M.; Zhu, K.; Fan, Q.; Li, J.; Ning, T.; Tian, F.; et al. Exosome circRNA secreted from adipocytes promotes the growth of hepatocellular carcinoma by targeting deubiquitination-related USP7. Oncogene 2019, 38, 2844–2859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Shojaei, S.; Hashemi, S.M.; Ghanbarian, H.; Sharifi, K.; Salehi, M.; Mohammadi-Yeganeh, S. Delivery of miR-381-3p Mimic by Mesenchymal Stem Cell-Derived Exosomes Inhibits Triple Negative Breast Cancer Aggressiveness; an In Vitro Study. Stem. Cell Rev. Rep. 2021, 17, 1027–1038. [Google Scholar] [CrossRef] [PubMed]
  94. Jia, Z.; Zhu, H.; Sun, H.; Hua, Y.; Zhang, G.; Jiang, J.; Wang, X. Adipose Mesenchymal Stem Cell-Derived Exosomal microRNA-1236 Reduces Resistance of Breast Cancer Cells to Cisplatin by Suppressing SLC9A1 and the Wnt/β-Catenin Signaling. Cancer Manag. Res. 2020, 12, 8733–8744. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, Y.; Chu, Y.; Li, K.; Zhang, G.; Guo, Z.; Wu, X.; Qiu, C.; Li, Y.; Wan, X.; Sui, J.; et al. Exosomes Secreted by Adipose-Derived Mesenchymal Stem Cells Foster Metastasis and Osteosarcoma Proliferation by Increasing COLGALT2 Expression. Front. Cell Dev. Biol. 2020, 8, 353. [Google Scholar] [CrossRef]
  96. Fan, X.; Wang, J.; Qin, T.; Zhang, Y.; Liu, W.; Jiang, K.; Huang, D. Exosome miR-27a-3p secreted from adipocytes targets ICOS to promote antitumor immunity in lung adenocarcinoma. Thorac. Cancer 2020, 11, 1453–1464. [Google Scholar] [CrossRef] [Green Version]
  97. Reza, A.; Choi, Y.J.; Yasuda, H.; Kim, J.H. Human adipose mesenchymal stem cell-derived exosomal-miRNAs are critical factors for inducing anti-proliferation signalling to A2780 and SKOV-3 ovarian cancer cells. Sci. Rep. 2016, 6, 38498. [Google Scholar] [CrossRef]
  98. Lin, R.; Wang, S.; Zhao, R.C. Exosomes from human adipose-derived mesenchymal stem cells promote migration through Wnt signaling pathway in a breast cancer cell model. Mol. Cell. Biochem. 2013, 383, 13–20. [Google Scholar] [CrossRef]
  99. Wu, Q.; Li, J.; Li, Z.; Sun, S.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Zhang, Y.; Sun, S.; et al. Exosomes from the tumour-adipocyte interplay stimulate beige/brown differentiation and reprogram metabolism in stromal adipocytes to promote tumour progression. J. Exp. Clin. Cancer Res. CR 2019, 38, 223. [Google Scholar] [CrossRef] [Green Version]
  100. Wu, Q.; Sun, S.; Li, Z.; Yang, Q.; Li, B.; Zhu, S.; Wang, L.; Wu, J.; Yuan, J.; Yang, C.; et al. Tumour-originated exosomal miR-155 triggers cancer-associated cachexia to promote tumour progression. Mol. Cancer 2018, 17, 155. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, H.; Zhu, L.; Bai, M.; Liu, Y.; Zhan, Y.; Deng, T.; Yang, H.; Sun, W.; Wang, X.; Zhu, K.; et al. Exosomal circRNA derived from gastric tumor promotes white adipose browning by targeting the miR-133/PRDM16 pathway. Int. J. Cancer 2019, 144, 2501–2515. [Google Scholar] [CrossRef] [PubMed]
  102. Hu, W.; Ru, Z.; Zhou, Y.; Xiao, W.; Sun, R.; Zhang, S.; Gao, Y.; Li, X.; Zhang, X.; Yang, H. Lung cancer-derived extracellular vesicles induced myotube atrophy and adipocyte lipolysis via the extracellular IL-6-mediated STAT3 pathway. Biochim. Et Biophys. Acta. Mol. Cell Biol. Lipids 2019, 1864, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  103. Hu, W.; Xiong, H.; Ru, Z.; Zhao, Y.; Zhou, Y.; Xie, K.; Xiao, W.; Xiong, Z.; Wang, C.; Yuan, C.; et al. Extracellular vesicles-released parathyroid hormone-related protein from Lewis lung carcinoma induces lipolysis and adipose tissue browning in cancer cachexia. Cell Death Dis. 2021, 12, 134. [Google Scholar] [CrossRef]
  104. Liu, A.; Pan, W.; Zhuang, S.; Tang, Y.; Zhang, H. Cancer cell-derived exosomal miR-425-3p induces white adipocyte atrophy. Adipocyte 2022, 11, 487–500. [Google Scholar] [CrossRef]
  105. Liu, Y.; Wang, M.; Deng, T.; Liu, R.; Ning, T.; Bai, M.; Ying, G.; Zhang, H.; Ba, Y. Exosomal miR-155 from gastric cancer induces cancer-associated cachexia by suppressing adipogenesis and promoting brown adipose differentiation via C/EPBβ. Cancer Biol. Med. 2022, 19, 9. [Google Scholar] [CrossRef]
  106. Ba, L.; Xue, C.; Li, X.; Zhang, M.; Yang, Y.; Han, Q.; Sun, Z.; Zhao, R.C. Gastric Cancer Cell-Derived Exosomes Can Regulate the Biological Functions of Mesenchymal Stem Cells by Inducing the Expression of Circular RNA circ_0004303. Stem. Cells Dev. 2021, 30, 830–842. [Google Scholar] [CrossRef] [PubMed]
  107. Di, W.; Zhang, W.; Zhu, B.; Li, X.; Tang, Q.; Zhou, Y. Colorectal cancer prompted adipose tissue browning and cancer cachexia through transferring exosomal miR-146b-5p. J. Cell. Physiol. 2021, 236, 5399–5410. [Google Scholar] [CrossRef]
  108. Luo, F.; Sun, Z.; Han, Q.; Xue, C.; Bai, C. Effect of Human Hepatocellular Carcinoma HepG2 Cell-derived Exosome on the Differentiation of Mesenchymal Stem Cells and Their Interaction. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. Acta Acad. Med. Sinicae 2017, 39, 312–317. [Google Scholar]
  109. Sagar, G.; Sah, R.P.; Javeed, N.; Dutta, S.K.; Smyrk, T.C.; Lau, J.S.; Giorgadze, N.; Tchkonia, T.; Kirkland, J.L.; Chari, S.T.; et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2016, 65, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  110. Milane, L.; Singh, A.; Mattheolabakis, G.; Suresh, M.; Amiji, M.M. Exosome mediated communication within the tumor microenvironment. J. Control. Release 2015, 219, 278–294. [Google Scholar] [CrossRef]
  111. Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Mohammadalipour, A.; Dumbali, S.P.; Wenzel, P.L. Mitochondrial Transfer and Regulators of Mesenchymal Stromal Cell Function and Therapeutic Efficacy. Front. Cell Dev. Biol. 2020, 8, 603292. [Google Scholar] [CrossRef] [PubMed]
  113. Wang, S.; Xu, M.; Xiao, X.; Wang, L.; Sun, Z.; Guan, M.; Zhao, R.C. Pancreatic cancer cell exosomes induce lipidomics changes in adipocytes. Adipocyte 2022, 11, 346–355. [Google Scholar] [CrossRef]
  114. Wang, S.; Xu, M.; Li, X.; Su, X.; Xiao, X.; Keating, A.; Zhao, R.C. Exosomes released by hepatocarcinoma cells endow adipocytes with tumor-promoting properties. J. Hematol. Oncol. 2018, 11, 1–14. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, X.; Li, Z.; Li, T.; Zhu, L.; Li, Z.; Tian, N. Long non-coding RNA HOTAIR enhances angiogenesis by induction of VEGFA expression in glioma cells and transmission to endothelial cells via glioma cell derived-extracellular vesicles. Am. J. Transl. Res. 2017, 9, 5012–5021. [Google Scholar] [PubMed]
  116. Zheng, Z.; Liu, L.; Zhan, Y.; Yu, S.; Kang, T. Adipose-derived stem cell-derived microvesicle-released miR-210 promoted proliferation, migration and invasion of endothelial cells by regulating RUNX3. Cell Cycle Georget. Tex. 2018, 17, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  117. Deng, Z.B.; Poliakov, A.; Hardy, R.W.; Clements, R.; Liu, C.; Liu, Y.; Wang, J.; Xiang, X.; Zhang, S.; Zhuang, X.; et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes 2009, 58, 2498–2505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. McNeal, J.E.; Villers, A.A.; Redwine, E.A.; Freiha, F.S.; Stamey, T.A. Capsular penetration in prostate cancer. Significance for natural history and treatment. Am. J. Surg. Pathol. 1990, 14, 240–247. [Google Scholar] [CrossRef]
  119. Takahara, K.; Ii, M.; Inamoto, T.; Nakagawa, T.; Ibuki, N.; Yoshikawa, Y.; Tsujino, T.; Uchimoto, T.; Saito, K.; Takai, T.; et al. microRNA-145 Mediates the Inhibitory Effect of Adipose Tissue-Derived Stromal Cells on Prostate Cancer. Stem. Cells Dev. 2016, 25, 1290–1298. [Google Scholar] [CrossRef]
  120. Rivera-Izquierdo, M.; Pérez de Rojas, J.; Martínez-Ruiz, V.; Arrabal-Polo, M.; Pérez-Gómez, B.; Jiménez-Moleón, J.J. Obesity and biochemical recurrence in clinically localised prostate cancer: A systematic review and meta-analysis of 86,490 patients. Prostate Cancer Prostatic Dis. 2022, 25, 411–421. [Google Scholar] [CrossRef]
  121. Kim, S.J.; Park, M.U.; Chae, H.K.; Nam, W.; Kim, S.W.; Yu, H.; Kim, H.G.; Kang, G.H.; Park, J.Y. Overweight and obesity as risk factors for biochemical recurrence of prostate cancer after radical prostatectomy. Int. J. Clin. Oncol. 2022, 27, 403–410. [Google Scholar] [CrossRef] [PubMed]
  122. Unamuno, X.; Gómez-Ambrosi, J.; Rodríguez, A.; Becerril, S.; Frühbeck, G.; Catalán, V. Adipokine dysregulation and adipose tissue inflammation in human obesity. Eur. J. Clin. Investig. 2018, 48, e12997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Marcelin, G.; Silveira, A.L.M.; Martins, L.B.; Ferreira, A.V.; Clément, K. Deciphering the cellular interplays underlying obesity-induced adipose tissue fibrosis. J. Clin. Investig. 2019, 129, 4032–4040. [Google Scholar] [CrossRef] [PubMed]
  124. Engin, A. Fat Cell and Fatty Acid Turnover in Obesity. Adv. Exp. Med. Biol. 2017, 960, 135–160. [Google Scholar] [PubMed]
  125. Durcin, M.; Fleury, A.; Taillebois, E.; Hilairet, G.; Krupova, Z.; Henry, C.; Truchet, S.; Trötzmüller, M.; Köfeler, H.; Mabilleau, G.; et al. Characterisation of adipocyte-derived extracellular vesicle subtypes identifies distinct protein and lipid signatures for large and small extracellular vesicles. J. Extracell. Vesicles 2017, 6, 1305677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hu, Q.; Su, H.; Li, J.; Lyon, C.; Tang, W.; Wan, M.; Hu, T.Y. Clinical applications of exosome membrane proteins. Precis. Clin. Med. 2020, 3, 54–66. [Google Scholar] [CrossRef]
  127. Jafari, N.; Kolla, M.; Meshulam, T.; Shafran, J.S.; Qiu, Y.; Casey, A.N.; Pompa, I.R.; Ennis, C.S.; Mazzeo, C.S.; Rabhi, N.; et al. Adipocyte-derived exosomes may promote breast cancer progression in type 2 diabetes. Sci. Signal. 2021, 14, eabj2807. [Google Scholar]
  128. Wen, Z.; Li, J.; Fu, Y.; Zheng, Y.; Ma, M.; Wang, C. Hypertrophic Adipocyte-Derived Exosomal miR-802-5p Contributes to Insulin Resistance in Cardiac Myocytes Through Targeting HSP60. Obes. Silver Spring Md. 2020, 28, 1932–1940. [Google Scholar] [CrossRef]
  129. Figueira, I.; Godinho-Pereira, J.; Galego, S.; Maia, J.; Haskó, J.; Molnár, K.; Malhó, R.; Costa-Silva, B.; Wilhelm, I.; Krizbai, I.A.; et al. MicroRNAs and Extracellular Vesicles as Distinctive Biomarkers of Precocious and Advanced Stages of Breast Cancer Brain Metastases Development. Int. J. Mol. Sci. 2021, 22, 5214. [Google Scholar] [CrossRef]
  130. Sano, S.; Izumi, Y.; Yamaguchi, T.; Yamazaki, T.; Tanaka, M.; Shiota, M.; Osada-Oka, M.; Nakamura, Y.; Wei, M.; Wanibuchi, H.; et al. Lipid synthesis is promoted by hypoxic adipocyte-derived exosomes in 3T3-L1 cells. Biochem. Biophys. Res. Commun. 2014, 445, 327–333. [Google Scholar] [CrossRef]
  131. Müller, G.; Schneider, M.; Biemer-Daub, G.; Wied, S. Microvesicles released from rat adipocytes and harboring glycosylphosphatidylinositol-anchored proteins transfer RNA stimulating lipid synthesis. Cell. Signal. 2011, 23, 1207–1223. [Google Scholar] [CrossRef] [PubMed]
  132. Iyengar, N.M.; Gucalp, A.; Dannenberg, A.J.; Hudis, C.A. Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J. Clin. Oncol. 2016, 34, 4270–4276. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, Y.; Daquinag, A.C.; Amaya-Manzanares, F.; Sirin, O.; Tseng, C.; Kolonin, M.G. Stromal progenitor cells from endogenous adipose tissue contribute to pericytes and adipocytes that populate the tumor microenvironment. Cancer Res. 2012, 72, 5198–5208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ribeiro, R.; Monteiro, C.; Catalán, V.; Hu, P.; Cunha, V.; Rodríguez, A.; Gómez-Ambrosi, J.; Fraga, A.; Príncipe, P.; Lobato, C.; et al. Obesity and prostate cancer: Gene expression signature of human periprostatic adipose tissue. BMC Med. 2012, 10, 108. [Google Scholar] [CrossRef] [Green Version]
  135. Hoy, A.J.; Balaban, S.; Saunders, D.N. Adipocyte–tumor cell metabolic crosstalk in breast cancer. Trends Mol. Med. 2017, 23, 381–392. [Google Scholar] [CrossRef]
  136. Incio, J.; Liu, H.; Suboj, P.; Chin, S.M.; Chen, I.X.; Pinter, M.; Ng, M.R.; Nia, H.T.; Grahovac, J.; Kao, S.; et al. Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. Cancer Discov. 2016, 6, 852–869. [Google Scholar] [CrossRef] [Green Version]
  137. Nead, K.T.; Sharp, S.J.; Thompson, D.J.; Painter, J.N.; Savage, D.B.; Semple, R.K.; Barker, A.; Perry, J.R.; Attia, J.; Dunning, A.M.; et al. Evidence of a Causal Association Between Insulinemia and Endometrial Cancer: A Mendelian Randomization Analysis. J. Natl. Cancer Inst. 2015, 107, djv178. [Google Scholar] [CrossRef]
  138. Pan, S.Y.; Johnson, K.C.; Ugnat, A.-M.; Wen, S.W.; Mao, Y. Association of obesity and cancer risk in Canada. Am. J. Epidemiol. 2004, 159, 259–268. [Google Scholar] [CrossRef] [Green Version]
  139. Bitting, R.L.; Armstrong, A.J. Targeting the PI3K/Akt/mTOR pathway in castration-resistant prostate cancer. Endocr. Relat. Cancer 2013, 20, R83–R99. [Google Scholar] [CrossRef] [Green Version]
  140. Marhold, M.; Tomasich, E.; El-Gazzar, A.; Heller, G.; Spittler, A.; Horvat, R.; Krainer, M.; Horak, P. HIF1α Regulates mTOR Signaling and Viability of Prostate Cancer Stem CellsHIF1α in Prostate Cancer Stem Cells. Mol. Cancer Res. 2015, 13, 556–564. [Google Scholar] [CrossRef] [Green Version]
  141. Ranasinghe, W.K.; Baldwin, G.S.; Shulkes, A.; Bolton, D.; Patel, O. Normoxic regulation of HIF-1α in prostate cancer. Nat Rev Urol. 2014, 11, 419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Fontana, F.; Anselmi, M.; Limonta, P. Molecular mechanisms and genetic alterations in prostate cancer: From diagnosis to targeted therapy. Cancer Lett. 2022, 534, 215619. [Google Scholar] [CrossRef] [PubMed]
  143. Tai, Y.L.; Chen, K.C.; Hsieh, J.T.; Shen, T.L. Exosomes in cancer development and clinical applications. Cancer Sci. 2018, 109, 2364–2374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wu, Q.; Li, B.; Li, Z.; Li, J.; Sun, S.; Sun, S. Cancer-associated adipocytes: Key players in breast cancer progression. J. Hematol. Oncol. 2019, 12, 95. [Google Scholar] [CrossRef] [PubMed]
  146. Rios-Colon, L.; Arthur, E.; Niture, S.; Qi, Q.; Moore, J.T.; Kumar, D. The Role of Exosomes in the Crosstalk between Adipocytes and Liver Cancer Cells. Cells 2020, 9, 1988. [Google Scholar] [CrossRef]
  147. Hui, L.; Chen, Y. Tumor microenvironment: Sanctuary of the devil. Cancer Lett. 2015, 368, 7–13. [Google Scholar] [CrossRef]
  148. Mohd Ali, N.; Yeap, S.K.; Ho, W.Y.; Boo, L.; Ky, H.; Satharasinghe, D.A.; Tan, S.W.; Cheong, S.K.; Huang, H.D.; Lan, K.C.; et al. Adipose MSCs Suppress MCF7 and MDA-MB-231 Breast Cancer Metastasis and EMT Pathways Leading to Dormancy via Exosomal-miRNAs Following Co-Culture Interaction. Pharm. Basel Switz. 2020, 14, 8. [Google Scholar] [CrossRef]
  149. Zhao, L.; Gu, C.; Gan, Y.; Shao, L.; Chen, H.; Zhu, H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J. Control. Release 2020, 318, 1–15. [Google Scholar] [CrossRef]
  150. Zhang, B.; Yin, Y.; Lai, R.C.; Lim, S.K. Immunotherapeutic potential of extracellular vesicles. Front. Immunol. 2014, 5, 518. [Google Scholar] [CrossRef] [Green Version]
  151. Pelosi, M.A., 3rd; Pelosi, M.A., 2nd. Liposuction. Obstet. Gynecol. Clin. N. Am. 2010, 37, 507–519. [Google Scholar] [CrossRef] [PubMed]
  152. Mead, B.; Tomarev, S. Bone Marrow-Derived Mesenchymal Stem Cells-Derived Exosomes Promote Survival of Retinal Ganglion Cells Through miRNA-Dependent Mechanisms. Stem. Cells Transl. Med. 2017, 6, 1273–1285. [Google Scholar] [CrossRef] [PubMed]
  153. Marote, A.; Teixeira, F.G.; Mendes-Pinheiro, B.; Salgado, A.J. MSCs-derived exosomes: Cell-secreted nanovesicles with regenerative potential. Front. Pharmacol. 2016, 7, 231. [Google Scholar] [CrossRef] [PubMed]
  154. Cho, B.S.; Lee, J.; Won, Y.; Duncan, D.I.; Jin, R.C.; Lee, J.; Kwon, H.H.; Park, G.-H.; Yang, S.H.; Park, B.C. Skin brightening efficacy of exosomes derived from human adipose tissue-derived stem/stromal cells: A prospective, split-face, randomized placebo-controlled study. Cosmetics 2020, 7, 90. [Google Scholar] [CrossRef]
Figure 1. PPAT in robot-assisted radical prostate cancer surgery. The orange color in the image represents PPAT and the blue color represents prostate cancer.
Figure 1. PPAT in robot-assisted radical prostate cancer surgery. The orange color in the image represents PPAT and the blue color represents prostate cancer.
Cancers 14 05077 g001
Figure 2. Potential role of exosomes in the intercommunication between PCa and PPAT in the obese state.
Figure 2. Potential role of exosomes in the intercommunication between PCa and PPAT in the obese state.
Cancers 14 05077 g002
Figure 3. A simple extraction protocol for adipose exosomes.
Figure 3. A simple extraction protocol for adipose exosomes.
Cancers 14 05077 g003
Table 1. Cross-talk between PPAT and PCa.
Table 1. Cross-talk between PPAT and PCa.
Type of ResearchTimesResearch SubjectsConclusionReferences
Clinical Research2011932 patients treated with brachytherapy or radiation therapy.There was an association between thickness and size of PPAT and high risk of PCa.[14]
Clinical Research2012652 prostate cancer patientsThe thickness of PPAT was associated with the detection rate of prostate cancer, especially high-grade PCa.[6]
Clinical Research2014184 patients who underwent radical retropubic prostatectomy.PPAT area and ratio (PPAT volume/prostate volume) were associated with high-risk PCa.[15]
Clinical Research2014308 patients treated with radiotherapy.PPAT regions were associated with PCade aggressiveness and were associated with skin color.[5]
Clinical Research2015190 PCa patients undergoing MRI.PPAT thickness was an independent predictor of PCa and high-grade PCa.[16]
Clinical Research2017371 patients with PCa, 292 patients with high-grade Pca.PPAT thickness was a potential detection metric for PCa and advanced PCa.[17]
Clinical Research2017162 patients who underwent MRI prior to prostatectomy.PPAT fat ratio correlated with PCa aggressiveness.[18]
Clinical Research2021175 prostate cancer patients (mean age 62.5 years, mean prostate-specific antigen 5.4 ng/dL).A higher periprostatic fat ratio was found to be significantly associated with a higher Gleason score by parametric magnetic resonance imaging (mpMRI).[19]
Clinical Research2021175 prostate cancer patients (mean age 62.5 years, mean prostate-specific antigen 5.4 ng/dL).Increased periprostatic fat volume was associated with disease progression in prostate cancer patients.[19]
Clinical Research202085 men with advanced PCa receiving ADT who had not received hormone therapy.PPAT thickness was a predictor of survival in patients with advanced PCa not receiving hormonal therapy.[20]
Basic Studies2012PPAT in prostate cancer patients, PC3, LNCaP.PPAT-derived factors increased migration of PC3 and LNCaP cell lines, while PPAT had a strong proliferative effect on PC3 cell lines.[21]
Basic Studies2021PPAT from 14 PCa patients (median age 62 years, median BMI 28.3) who underwent radical prostatectomy, DU145, PC3Conditioned medium (CM) culture of PPAT promoted migration of human androgen non-dependent PCa cell lines and upregulated CTGF expression.[22]
Basic Studies2018DU145, PPAT in 36 Caucasians and 36 African-CaribbeansFatty acid (FA) content in PPAT is associated with PCa progression.[23]
Basic Studies2021PPAT in vitro culture collection of conditioned medium, DU145, PC3.PPAT secreted IGF-1 to upregulate TUBB2B β-microtubulin heterodimer to promote resistance to doxorubicin in prostate cancer.[24]
Basic Studies2012PPAT, PC-3, and LNCaP cell lines from prostate cancer patients.PPAT increased MMP (matrix metalloproteinase) activity to regulate the microenvironment of extraprostatic tumor cells and promoted prostate cancer cell survival and migration.[21]
Basic Studies2019Prostate cancer cell lines C4-2B, Du-145, and PC-3.Free fatty acids released by PPAT promoted tumor progression by affecting the HIF1/MMP14 pathway by stimulating NOX5/ROS.[25]
Basic Studies2018Primary NK cells, C4-2, 3T3-L1.Inhibition of the IL-6/leptin-JAK/Stat3 signaling axis in adipocytes enhanced immune killing of CRPC (castration-resistant prostate cancer) cells by NK cells.[26]
Basic Studies2021Adipocytes isolated from PCa patients and PC3, 22RV1 Decreased autophagic activity and increased intracellular lipid droplet content in PC3 cells after co-culture with adipocytes.[27]
Basic Studies2012PPAT, LNCaP, PC3 in PCa patients.PPAT-released pro-MMP-9 induced invasiveness of LNCaP (androgen-dependent) cells.[28]
Basic Studies2009PPAT collected from patients undergoing radical prostatectomy.PPAT regulated the aggressiveness of prostate cancer by providing IL-6.[13]
Table 2. Tumorigenic effects of adipocyte-derived exosomes.
Table 2. Tumorigenic effects of adipocyte-derived exosomes.
Exosome CargoSourceRole in TumorReferences
MMP33T3-L1 adipocytesInduction of lung cancer metastasis through activation of the MMP3/MMP9 process.[87]
miRNA-21Cancer-associated adipocytesInhibition of the apoptotic process in ovarian cancer cells.[89]
-3T3-L1 adipocytesReducing degradation of caspase 3/PARP molecules in PCa and improving resistance to doxorubicin in prostate cancer.[37]
-Adipocytes in the obese stateEnhanced estrogen receptor expression and growth, motility and invasion, stem-cell-like properties and epithelial–mesenchymal transition of triple-negative breast cancer cells through induction of HIF-1α activity.[90]
miR-3940-5p, miR-22-3p, miR-16-5pAdipose mesenchymal stem cellsInhibiting the proliferation and migration of rectal cancer.[91]
circ-DB3T3-L1 adipocytesInhibiting miR-34a and activating USP7/Cyclin A2 signaling pathway promote hepatocellular carcinoma growth and reduce DNA damage.[92]
miR-381-3pAdipose mesenchymal stem cellsInhibition of apoptosis and progression of triple-negative breast cancer cells.[93]
microRNA-1236Adipose mesenchymal stem cellsInhibiting SLC9A1 and Wnt/β-linked protein signaling to reduce cisplatin resistance in breast cancer cells.[94]
-Adipose mesenchymal stem cellsIncreasing COLGALT2 expression to promote osteosarcoma proliferation and metastasis.[95]
miR-27a-3p3T3-L1 adipocytesInhibiting ICOS+ T cell proliferation and IFN-γ secretion to alter the immune microenvironment of lung adenocarcinoma.[96]
miR-23a/b3T3-L1 adipocytesTargeting the VHL/HIF axis to promote HCC cell growth and migration.[88]
hsa-miR-124-3pAdipose mesenchymal stem cellsInhibiting the growth and proliferation of ovarian cancer cells.[97]
microRNA-21Cancer-associated adipocytesTargeting APAF1 promotes paclitaxel resistance in ovarian cancer cells.[89]
-Adipose mesenchymal stem cellsMediated Wnt signaling pathway induces migration of breast cancer cells.[98]
Table 3. Effects of tumor-derived exosomes on adiposity.
Table 3. Effects of tumor-derived exosomes on adiposity.
Exosome CargoSourceRole in AdiposeReferences
miRNA-126Breast cancerDecreases the uptake of glucose by fat cells and increases their secretion of lactate and pyruvate.[99]
miRNA-155Breast cancerPromotes beige/brown differentiation and remodeling of adipocytes through downregulation of PPARγ expression.[100]
ciRS-133Gastric cancerRegulating preadipocytes and regulating preadipocyte differentiation.[101]
IL-6Lung cancerInducing adipocyte lipolysis by mediating the STAT3 pathway.[102]
Parathyroid hormone-related proteinLewis lung carcinomaInducting lipolysis and adipose tissue browning through the PKA pathway.[103]
miR-425-3pA549, H1299 and AGSInducing white adipocyte atrophy.[104]
miR-155Stomach cancer cellsInhibiting adipogenesis and promoting brown adipose differentiation via C/EPBβ pathway in adipose mesenchymal stem cells.[105]
ciRS-133Gastric cancer cellsActivation of PRDM16 and inhibition of miR-133 promote differentiation of preadipocytes into brown adipocytes.[101]
circ_0004303Gastric cancer cellsPromoting migration and invasion of adipose mesenchymal stem cells.[106]
miR-146b-5pHuman colorectal cancer tissuePromoting adipose tissue browning and inhibiting HOXC10 to accelerate lipolysis.[107]
-HepG2Inducing adipose MSCs to differentiate into cancer-associated myofibroblasts.[108]
AdrenomedullinHuman pancreatic cancer tissue exosomesActivating p38 and ERK1/2 MAPK and promoting lipolysis by phosphorylating hormone-sensitive lipase.[109]
H-ras, miR-125b, miR-155, and GTPasesC4-2B prostate cancer cellsInducing prostate-tumor-like transformation of adipose stem cells.[44]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Feng, S.; Lou, K.; Luo, C.; Zou, J.; Zou, X.; Zhang, G. Obesity-Related Cross-Talk between Prostate Cancer and Peripheral Fat: Potential Role of Exosomes. Cancers 2022, 14, 5077. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14205077

AMA Style

Feng S, Lou K, Luo C, Zou J, Zou X, Zhang G. Obesity-Related Cross-Talk between Prostate Cancer and Peripheral Fat: Potential Role of Exosomes. Cancers. 2022; 14(20):5077. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14205077

Chicago/Turabian Style

Feng, Shangzhi, Kecheng Lou, Cong Luo, Junrong Zou, Xiaofeng Zou, and Guoxi Zhang. 2022. "Obesity-Related Cross-Talk between Prostate Cancer and Peripheral Fat: Potential Role of Exosomes" Cancers 14, no. 20: 5077. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14205077

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop