Lactate/GPR81 signaling and proton motive force in cancer: Role in angiogenesis, immune escape, nutrition, and Warburg phenomenon

https://doi.org/10.1016/j.pharmthera.2019.107451Get rights and content

Abstract

Reprogramming of biochemical pathways is a hallmark of cancer cells, and generation of lactic acid from glucose/glutamine represents one of the consequences of such metabolic alterations. Cancer cells export lactic acid out to prevent intracellular acidification, not only increasing lactate levels but also creating an acidic pH in extracellular milieu. Lactate and protons in tumor microenvironment are not innocuous bystander metabolites but have special roles in promoting tumor-cell proliferation and growth. Lactate functions as a signaling molecule by serving as an agonist for the G-protein-coupled receptor GPR81, involving both autocrine and paracrine mechanisms. In the autocrine pathway, cancer cell-generated lactate activates GPR81 on cancer cells; in the paracrine pathway, cancer cell-generated lactate activates GPR81 on immune cells, endothelial cells, and adipocytes present in tumor stroma. The end result of GPR81 activation is promotion of angiogenesis, immune evasion, and chemoresistance. The acidic pH creates an inwardly directed proton gradient across the cancer-cell plasma membrane, which provides driving force for proton-coupled transporters in cancer cells to enhance supply of selective nutrients. There are several molecular targets in the pathways involved in the generation of lactic acid by cancer cells and its role in tumor promotion for potential development of novel anticancer therapeutics.

Introduction

Otto Warburg's findings from the 1920s highlighted the preferential production of lactic acid in glycolysis even in the presence of oxygen (Warburg, 1956). These were unexpected findings because in normal cells glycolysis produces pyruvic acid which enters mitochondria and undergoes complete oxidation into CO2 in the presence of oxygen with consequent production of ATP via oxidative phosphorylation (OXPHOS). ATP is a potent negative feedback regulator of glycolysis via inhibition of the rate-limiting enzyme in the glycolytic pathway, namely phosphofructokinase-1; therefore, oxygen suppresses the rate of glycolysis in normal cells. In general, glycolysis yields lactic acid in normal cells only under conditions of hypoxia. As lactic acid is produced in normal cells only in the absence of oxygen, the process is referred to as “anaerobic glycolysis”, and the altered pathway seen in cancer cells where lactic acid is produced even in the presence of oxygen is now referred to as “aerobic glycolysis”. Warburg proposed that this altered metabolism constitutes the origin of cancer (Warburg, 1956). But this novel theory of cancer metabolism did not receive much attention in Warburg's days nor the decades following; Warburg's findings in cancer cells were generally thought to represent a symptom of cancer rather than the primary cause. This notion has changed dramatically in the past two decades, and we are seeing a rekindling of interest in recent years in cancer-cell metabolism. Notwithstanding this increased interest in cancer-cell metabolism because of the original Warburg's findings, it has to be borne in mind that “aerobic glycolysis” is a phenomenon observed in cancer cells in culture. The extent to which this phenomenon occursin tumors in vivo is not clear. Nonetheless, the fact remains that there has certainly been an increased focus not only on glycolysis as it pertains directly to Warburg's original findings but also on the citric acid cycle, electron transport chain, oxidative phosphorylation, hexose monophosphate shunt, fatty acid metabolism, and amino acid metabolism in relation to tumor growth and metastasis.

The present review focuses on biologic and metabolic relevance of lactic acid generated by cancer cells, highlighting the notion that this glycolytic metabolite is not a waste product of proliferating cancer cells but actually elicits a broad spectrum of effects that are critical for tumor progression and metastasis. To avoid intracellular acidification, cancer cells rapidly export lactate via monocarboxylate transporters (MCTs) (Ganapathy, Thangaraju, & Prasad, 2009; Parks & Pouysségur, 2017). This maintains low intracellular concentrations of lactic acid and allows for aerobic glycolysis and lactate production to continue, and simultaneously increases lactic acid concentrations in the extracellular tumor microenvironment (TME). The pKa of lactic acid is ~3.9; therefore, lactic acid exists mostly in the ionized state at physiological pH. Thus, lactic acid has two components, lactate and H+; tumor-promoting effects of lactic acid involve both components. In addition to the role of lactate as an energy-rich metabolite, lactate has now emerged as an important signaling molecule acting through the cell-surface Gi/o-protein-coupled receptor GPR81 (also known as hydroxycarboxylic acid receptor 1 or HCAR1). High lactate concentrations in the extracellular environment of tumors is associated with a poor prognosis (Sun, Li, Chen, & Qian, 2017). Accumulation of lactic acid in the TME acidifies the extracellular milieu due to H+, thus creating a transmembrane electrochemical H+ gradient (also known as proton motive force) across the plasma membrane of cancer cells. As the proton motive force provides energy for active entry of several essential nutrients (amino acids, small peptides, folic acid, and Fe2+) in normal cells, a similar phenomenon might occur also in cancer cells. In the current review, we will detail the biological roles of both lactate and H+ in cancer cells and in the TME as they pertain to cancer progression and metastasis.

Section snippets

Lactic acid as a cancer cell-specific metabolite and its connection to Warburg effect

Otto Warburg found the rate of glycolysis to be much higher in cancer cells than in normal cells, and that pyruvic acid was preferentially converted to lactic acid as the end product of glycolysis even in the presence of oxygen. The conversion of pyruvic acid to lactic acid is obligatory for sustaining increased rates of glycolysis in cancer cells (Fig. 1). The basis of this functional coupling is the fact that the continued activity of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) that

Role of glutaminolysis in the generation of lactic acid in cancer cells

In addition to aerobic glycolysis, tumor-derived lactic acid is also generated from catabolism of glutamine (DeBerardinis et al., 2007). Coordinated by c-Myc, cancer cells induce selective amino acid transporters to increase glutamine uptake from their surroundings (Bhutia, Babu, Ramachandran, & Ganapathy, 2015; Bhutia & Ganapathy, 2016). In addition, glutaminase is induced to facilitate the conversion of glutamine to glutamate, whereupon intracellular glutamine can enter the citric acid cycle

Plasma membrane transporters (MCT1 and MCT4) for the efflux/influx of lactic acid in cancer cells

Monocarboxylate transporters (MCTs) are members of the SLC16 gene family which facilitate bidirectional symport of monocarboxylates and H+ across cell membranes (Halestrap, 2013). MCTs 1–4 are the major physiologically relevant isoforms that function in the H+-coupled transport of molecules containing a single carboxylate group such as pyruvate, lactate, acetoacetate and β-hydroxybutyrate. MCT1 (SLC16A1) and MCT4 (SLC16A3) are ubiquitously expressed in humans, and highly upregulated in cancers

Lactate as a source of metabolic energy for cancer cells in an oxygen-rich environment: metabolic symbiosis

Lactate shuttling between different cell populations within the TME has emerged as a new phenomenon in the field of tumor biology. Lactate shuttling encompasses many physiological and pathological scenarios where lactate is exported by one cell type and imported by another cell type. The Cori cycle involving the lactate shuttle between skeletal muscle and liver is well known. Because lactate is an energy-rich metabolite capable of serving as a precursor for both gluconeogenesis and ATP

Cancer-associated fibroblasts as lactate producers: reverse Warburg effect

Metabolic symbiosis occurs not only between different types of cancer cells within the tumor but also between normoxic cancer cells and tumor-associated stromal cells, particularly fibroblasts (Martinez-Outschoom, Lisanti, & Sotgia, 2014; Martinez-Outschoorn, Sotgia, & Lisanti, 2014). Cancer cells apparently “instruct” cancer-associated fibroblasts to switch to an “aerobic glycolysis” metabolic phenotype and transform into lactate producers within the TME. These fibroblasts are deficient in

Lactic acid and tumor angiogenesis: intracellular actions

Angiogenesis and high lactate concentrations are both critical components of healing wounds. Lactic acid accumulation in the heterogeneous wound microenvironment is a result of both aerobic and anerobic glycolysis by hypoxic and damaged cells, immune-cell activation, and cell proliferation. In a sense, the TME shares many similarities with the wound microenvironment. Wound healing involves migration of macrophages and fibroblasts which rely on glycolytic substrate-level phosphorylation for ATP

Lactic acid and tumor immunology

Immune cells comprise an important component of the TME, and no different than cancer cells, immune cells have a diverse metabolic portfolio with regard to lactic acid flux. Lactic acid impacts tumor growth by acting as both a pro-inflammatory and immunosuppressive molecule. However, lactic acid predominately dampens the immune response to cancer, and overall there are both H+-dependent and lactate-dependent mechanisms by which lactic acid can enable tumors to evade immune-mediated destruction.

Additional mechanisms for tumor promotion by lactic acid

Glutamine is another major source of lactic acid production in the TME. In addition to activating HIF-1α and NF-kB, lactate signaling has also been implicated in hypoxia-independent activation of c-Myc via post-translational stabilization of HIF-2α (Pérez-Escuredo et al., 2016). Pyruvate generated from MCT1-imported lactate stabilizes HIF-2α which then activates the c-Myc transcriptional complex. In turn, activated c-Myc upregulates expression of the glutamine transporter ASCT2 and the

Lactate as an extracellular messenger: involvement of GPR81

The emergence of lactate as a signaling molecule has come full circle with the discovery of the lactate receptor GPR81 in the plasma membrane of adipocytes (Ge et al., 2008). GPR81 expression is also found in muscle cells, the central nervous system, immune cells, and now tumor cells. Lactate signaling via GPR81 is especially unique from everything we have discussed up to this point in that signaling through GPR81 does not require H+ or MCT-driven import nor does it require conversion to

Biological functions of GPR81 in normal tissues

GPR81 belongs to a subfamily of GPCRs, termed the hydroxy carboxylic acid receptors (HCARs), consisting of three members: HCAR1 (GPR81), HCAR2 (GPR109A), and HCAR3 (GPR109B) (Offermanns et al., 2011). This unique family of GPCRs is localized sequentially on chromosome 12q in humans. In rodents, only Hcar1 and 2 are expressed, both located on chromosome 5 in mouse. Hcar3 is absent in rodents. HCAR1 is believed to be the most evolutionarily conserved of the three receptors, in that GPR81 is found

Relevance of GPR81 to inflammation

Several studies have investigated the role of lactate via GPR81 in infection and inflammation. GPR81 expression in adipocytes and endothelial cells of the blood-brain barrier are reduced under inflammatory conditions while GPR81 expression in immune cells is associated with protection from inflammation, and down-regulation of innate immunity (Boitsova et al., 2018; Feingold, Moser, Shigenaga, & Grunfeld, 2011; Ranganathan et al., 2018). Perhaps most intriguing was the role of GPR81 in the

Expression of GPR81 in cancer cells: Autocrine functions of lactate

As the cell-surface receptor for lactate, GPR81 might have an important role in tumor growth based on the fact that cancer cells generate lactic acid as the end product of glycolysis and export it out to the extracellular milieu. This role could be autocrine where extracellular lactate acts on GPR81 expressed on tumor cells themselves (Fig. 5). Alternatively, it could be paracrine where extracellular lactate produced by cancer cells act on GPR81 expressed on non-cancer cells in the TME (Fig. 6

Expression of GPR81 in non-tumor cells in tumor microenvironment: paracrine functions of lactate

More recently, our group and others have identified Gpr81 in immune cells (Brown, Ramachandran, Offermanns, & Ganapathy, 2019; Hoque et al., 2014; Ranganathan et al., 2018). Specifically, Gpr81 is expressed in dendritic cells and macrophages in the colon where lactate binding helps confer an immunosuppressive and protective phenotype (Ranganathan et al., 2018). While lactate/Gpr81-induced immunosuppression is protective in the colon, a similar function in the TME would be detrimental to the

Acidic pH in tumor microenvironment and the resultant proton motive force

Lactic acid generated via aerobic glycolysis in cancer cells dissociates into lactate and H+ under conditions of intracellular pH. The monocarboxylate transporters MCT1/4 mediate the efflux of lactic acid by facilitating the symport of lactate and H+. Thus, when lactic acid effluxes out of cancer cells, intracellular acidification is prevented and the extracellular milieu is acidified. Therefore, MCT1/4-mediated lactic acid efflux makes at least a partial contribution to the acidic TME. In

Transporters driven by proton motive force and their relevance to tumor-cell nutrition

The notably high rate of proliferation in cancer cells is obligatorily dependent on optimal availability of nutrients to support their energy demands and anabolic pathways. As such, cancer cells upregulate selective nutrient transporters to satisfy this increased need for nutrients (Bhutia et al., 2015; Bhutia & Ganapathy, 2016; Ganapathy et al., 2009). Perhaps the most recognized example is the facilitative glucose transporter GLUT1 (SLC2A1) whose upregulation in cancer is supported by c-Myc

Conclusions and perspectives

The preferential generation of lactic acid as the end product of glycolysis in cancer cells and the impact of this metabolite on tumor growth and metastasis have been recognized for several decades. The metabolic pathways associated with this phenomenon have highlighted several new drug targets for cancer treatment; this includes LDHA that converts pyruvate to lactate in cancer cells, and the monocarboxylate transporters MCT1 and MCT4 that mediate the influx and efflux of lactic acid in

Declaration of Competing Interest

The authors declare that there are no conflicts of interest.

Acknowledgments

This work was supported by the National Institutes of Health grant CA190710 and the Welch Endowed Chair in Biochemistry, Grant No. BI-0028, at Texas Tech University Health Sciences Center.

References (150)

  • M.E. Ganapathy et al.

    Differential recognition of beta -lactam antibiotics by intestinal and renal peptide transporters, PEPT 1 and PEPT 2

    The Journal of Biological Chemistry

    (1995)
  • M.E. Ganapathy et al.

    Valacyclovir: A substrate for the intestinal and renal peptide transporters PEPT1 and PEPT2

    Biochemical and Biophysical Research Communications

    (1998)
  • V. Ganapathy et al.

    Nutrient transporters in cancer: Relevance to Warburg hypothesis and beyond

    Pharmacology & Therapeutics

    (2009)
  • H. Ge et al.

    Elucidation of signaling and functional activities of an orphan GPCR, GPR81

    Journal of Lipid Research

    (2008)
  • E. Gopal et al.

    Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney

    Biochimica et Biophysica Acta

    (2007)
  • A.P. Halestrap

    The SLC16 gene family – Structure, role and regulation in health and disease

    Molecular Aspects of Medicine

    (2013)
  • R. Hoque et al.

    Lactate reduces liver and pancreatic injury in toll-like receptor- and inflammasome-mediated inflammation via GPR81-mediated suppression of innate immunity

    Gastroenterology

    (2014)
  • L. Ippolito et al.

    Lactate: A metabolic driver in the tumour landscape

    Trends in Biochemical Sciences

    (2019)
  • K. Kunzelmann et al.

    Bicarbonate in cystic fibrosis

    Journal of Cystic Fibrosis

    (2017)
  • H.W. Lee et al.

    Clinicopathologic significance of G protein-coupled receptor 81 in hepatocellular carcinoma

    International Journal of Cancer Research and Therapeutics

    (2017)
  • D.C. Lee et al.

    A lactate-induced response to hypoxia

    Cell

    (2015)
  • C. Liu et al.

    Lactate inhibits lipolysis in fat cells through activation of an orphan G-protein-coupled receptor, GPR81

    The Journal of Biological Chemistry

    (2009)
  • H. Lu et al.

    Reversible inactivation of HIF-1 prolyl hydroxylases allows cell metabolism to control basal HIF-1

    The Journal of Biological Chemistry

    (2005)
  • A. Madaan et al.

    Müller cell-localized G-protein-coupled receptor 81 (Hydroxycarboxylic Acid Receptor 1) regulates inner retinal vasculature via norrin/Wnt pathways

    The American Journal of Pathology

    (2019)
  • A. Madaan et al.

    Lactate produced during labor modulates uterine inflammation via GPR81 (HCA1)

    American Journal of Obstetrics and Gynecology

    (2017)
  • U. Martinez-Outschoorn et al.

    Tumor microenvironment and metabolic synergy in breast cancers: Critical importance of mitochondrial fuels and function

    Seminars in Oncology

    (2014)
  • K. Mitsuoka et al.

    Inhibition of oligopeptide transporter suppress growth of human pancreatic cancer cells

    European Journal of Pharmaceutical Sciences

    (2010)
  • S. Miyauchi et al.

    Functional identification of SLC5A8, a tumor suppressor down-regulated in colon cancer, as a Na+-coupled transporter for short-chain fatty acids

    The Journal of Biological Chemistry

    (2004)
  • S.K. Parks et al.

    Targeting pH regulating proteins for cancer therapy-Progress and limitations

    Seminars in Cancer Biology

    (2017)
  • J. Perez-Escuredo et al.

    Monocarboxylate transporters in the brain and in cancer

    Biochimica et Biophysica Acta

    (2016)
  • K. Peter et al.

    Lactic acid delays the inflammatory response of human monocytes

    Biochemical and Biophysical Research Communications

    (2015)
  • A.P. Andersen et al.

    The net acid extruders NHE1, NBCn1 and MCT4 promote mammary tumor growth through distinct but overlapping mechanisms

    International Journal of Cancer

    (2018)
  • C.M. Anderson et al.

    Transport of the photodynamic therapy agents 5-aminolevulinic acid by distinct H+-coupled nutrient carriers co-expressed in the small intestine

    The Journal of Pharmacology and Experimental Therapeutics

    (2010)
  • A. Angelin et al.

    FoxP3 reprograms T cell metabolism to function in low-glucose, high-lactate environments

    Cell Metabolism

    (2017)
  • M. Apicella et al.

    Increased lactate secretion by cancer cells sustains non-cell-autonomous adaptive resistance to MET and EGFR targeted therapies

    Cell Metabolism

    (2018)
  • F. Baumann et al.

    Lactate promotes glioma migration by TGF-beta2-dependent regulation of matrix metalloproteinase-2

    Neurooncology

    (2009)
  • D. Benjamin et al.

    Dual inhibition of the lactate transporters MCT1 and MCT4 is synthetic lethal with metformin due to NAD+ depletion in cancer cells

    Cell Reports

    (2018)
  • Y.D. Bhutia et al.

    Amino acid transporters in cancer and their relevance to “glutamine addiction”: Novel targets for the design of a new class of anticancer drugs

    Cancer Research

    (2015)
  • Y.D. Bhutia et al.

    SLC transporters as a novel class of tumour suppressors: Identity, function and molecular mechanisms

    The Biochemical Journal

    (2016)
  • E. Boedtkjer

    Na+, HCO3 cotransporter NBCn1 accelerates breast carcinogenesis

    Cancer Metastasis Reviews

    (2019)
  • E.B. Boitsova et al.

    The inhibitory effect of LPS on the expression of GPR81 lactate receptor in blood-brain barrier model in vitro

    Journal of Neuroinflammation

    (2018)
  • D. Borowitz

    CFTR, bicarbonate, and the pathophysiology of cystic fibrosis

    Pediatric Pulmonology

    (2015)
  • J. Boult et al.

    Overexpression of cellular iron import proteins is associated with malignant progression of esophageal adenocarcinoma

    Clinical Cancer Research

    (2008)
  • L. Bozzo et al.

    Lactate modulates the activity of primary cortical neurons through a receptor-mediated pathway

    PLoS One

    (2013)
  • T. Brown et al.

    The lactate receptor Gpr81 on non-cancer cells promotes an immunosuppressive phenotype in the tumor microenvironment

    Cancer Research

    (2019)
  • R.A. Cairns et al.

    Metabolic targeting of hypoxia and HIF1 in solid tumors can enhance cytotoxic chemotherapy

    Proceedings of the National Academy of Sciences of the United States of America

    (2007)
  • A. Calcinotto et al.

    Modulation of microenvironment acidity reverses anergy in human and murine tumor-infiltrating T lymphocytes

    Cancer Research

    (2012)
  • E. Calzoni et al.

    Biocompatible polymer nanoparticles for drug delivery applications in cancer and neurodegenerative disorder therapies

    J. Funct. Biomater.

    (2019)
  • X. Castillo et al.

    A probable dual mode of action for both L- and D-lactate neuroprotection in cerebral ischemia

    Journal of Cerebral Blood Flow and Metabolism

    (2015)
  • D. Chiabrando et al.

    Heme in pathophysiology: A matter of scavenging, metabolism and trafficking across cell membranes

    Frontiers in Pharmacology

    (2014)
  • Cited by (178)

    • pH regulators and their inhibitors in tumor microenvironment

      2024, European Journal of Medicinal Chemistry
    View all citing articles on Scopus
    View full text